151
|
van den Bos C, Keefe R, Schirmaier C, McCaman M. Therapeutic human cells: manufacture for cell therapy/regenerative medicine. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 138:61-97. [PMID: 23934363 DOI: 10.1007/10_2013_233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
: Human primary cells (e.g. adult stem cells) as well as differentiated cells, including those of the immune system, have been found to be therapeutically useful and free of ethical concerns. Several products have received market authorization and numerous promising clinical trials are underway. We believe that such primary therapeutic cells will dominate the market for cell therapy applications for the foreseeable future. Consequently, production of such cellular products warrants attention and needs to be a fully controlled pharmaceutical process. Thus, where possible, such production should change from manufacture towards a truly scalable industrialized process for both allogeneic and autologous products. Here, we discuss manufacturing aspects of both autogeneic and allogeneic products, review the field, and provide historical context.
Collapse
|
152
|
Rojas JJ, Sampath P, Bonilla B, Ashley A, Hou W, Byrd D, Thorne SH. Manipulating TLR Signaling Increases the Anti-tumor T Cell Response Induced by Viral Cancer Therapies. Cell Rep 2016; 15:264-73. [PMID: 27050526 PMCID: PMC4830920 DOI: 10.1016/j.celrep.2016.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/08/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022] Open
Abstract
The immune response plays a key role in enhancing the therapeutic activity of oncolytic virotherapies. However, to date, investigators have relied on inherent interactions between the virus and the immune system, often coupled to the expression of a single cytokine transgene. Recently, the importance of TLR activation in mediating adaptive immunity has been demonstrated. We therefore sought to influence the type and level of immune response raised after oncolytic vaccinia therapy through manipulation of TLR signaling. Vaccinia naturally activates TLR2, associated with an antibody response, whereas a CTL response is associated with TLR3-TRIF-signaling pathways. We manipulated TLR signaling by vaccinia through deglycosylation of the viral particle to block TLR2 activation and expression of a TRIF transgene. The resulting vector displayed greatly reduced production of anti-viral neutralizing antibody as well as an increased anti-tumor CTL response. Delivery in both naive and pre-treated mice was enhanced and immunotherapeutic activity dramatically improved.
Collapse
Affiliation(s)
- Juan J Rojas
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Padma Sampath
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Braulio Bonilla
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexandra Ashley
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Weizhou Hou
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Daniel Byrd
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Steve H Thorne
- Department of Cell Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
153
|
Ren R, Koti M, Hamilton T, Graham CH, Nayak JG, Singh J, Drachenberg DE, Siemens DR. A primer on tumour immunology and prostate cancer immunotherapy. Can Urol Assoc J 2016; 10:60-5. [PMID: 26977209 DOI: 10.5489/cuaj.3418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
No abstract
Collapse
Affiliation(s)
- Runhan Ren
- Department of Urology, Queen's University, Kingston, ON, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Thomas Hamilton
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Charles H Graham
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jasmir G Nayak
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | - Jas Singh
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | | | - D Robert Siemens
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
154
|
T Cells Derived From Human Melanoma Draining Lymph Nodes Mediate Melanoma-specific Antitumor Responses In Vitro and In Vivo in Human Melanoma Xenograft Model. J Immunother 2016; 38:229-38. [PMID: 26049546 DOI: 10.1097/cji.0000000000000078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
It has been established in murine models that lymph nodes draining a progressively growing tumor contain antigen-specific T cells capable of mediating protective immune responses upon adoptive transfer. However, naturally occurring human tumor-draining lymph nodes (TDLNs) have yet to be fully investigated. In this study, we analyzed TDLNs from patients with stage III melanoma who were undergoing routine lymph node dissection. Following short-term (14 d) culture activation with anti-CD3/anti-CD28 microbeads and expansion in low concentrations of IL-2, the melanoma-draining lymph node (MDLN) cells were ∼ 60% CD4-activated and ∼ 40% CD8-activated T cells. The activated MDLN cells demonstrated reactivity in response to overlapping peptides spanning the sequence of 4 different known melanoma antigens MAGEA1, Melan-A/MART-1, NY-ESO-1, and Prame/OIP4, suggesting the presence of melanoma-specific T cells. Coculture of activated MDLN T cells with cancer cells in vitro resulted in preferential apoptosis of human cancer cell lines that were cocultured with T cells with high degree of MHC matching. Adoptive transfer of MDLN T cells with high degree of MHC matching to A375 to mice-bearing human A375 melanoma xenografts resulted in dose-dependent improvement in survival. Although prior human studies have demonstrated the immune responses within melanoma vaccine-draining lymph nodes, this study presents evidence for the first time that naturally occurring human MDLN samples contain melanoma-experienced CD4 and CD8 T cells that can be readily cultured and expanded to mediate protective immune responses both in vitro and in vivo in a human melanoma xenograft model.
Collapse
|
155
|
Echarri MJ, Lopez-Martin A, Hitt R. Targeted Therapy in Locally Advanced and Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma (LA-R/M HNSCC). Cancers (Basel) 2016; 8:cancers8030027. [PMID: 26927178 PMCID: PMC4810111 DOI: 10.3390/cancers8030027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 01/07/2023] Open
Abstract
Surgery and radiotherapy are the standard treatment options for patients with squamous cell carcinoma of the head and neck (SCCHN). Chemoradiotherapy is an alternative for patients with locally advanced disease. In recurrent/metastatic disease and after progression to platin-based regimens, no standard treatments other than best supportive care are currently available. Most SCCHN tumours overexpress the epidermal growth factor receptor (EGFR). This receptor is a tyrosine-kinase membrane receptor that has been implicated in angiogenesis, tumour progression and resistance to different cancer treatments. In this review, we analysed the different drugs and pathways under development to treat SCCHN, especially recurrent/metastatic disease. Until now, the EGFR signalling pathway has been considered the most important target with respect to new drugs; however, new drugs, such as immunotherapies, are currently under study. As new treatments for SCCHN are developed, the influence of therapies with respect to overall survival, progression free survival and quality of life in patients with this disease is changing.
Collapse
Affiliation(s)
- María José Echarri
- Department of Medical Oncology, Hospital Universitario Severo Ochoa, Avenida Orellana s/n, Leganés, 28911 Madrid, Spain.
| | - Ana Lopez-Martin
- Department of Medical Oncology, Hospital Universitario Severo Ochoa, Avenida Orellana s/n, Leganés, 28911 Madrid, Spain.
| | - Ricardo Hitt
- Department of Medical Oncology, Hospital Universitario Severo Ochoa, Avenida Orellana s/n, Leganés, 28911 Madrid, Spain.
| |
Collapse
|
156
|
Abstract
Vaccination started around the 10th century AD as a means of preventing smallpox. By the end of the 19th century such therapeutic vaccines were well established with both active and passive preparations being used in clinical practice. Active immunization involved administering an immunogen that might be live/ attenuated, killed/ inactivated, toxoid or subunit in origin. Passive immunization involved giving pre-formed antibodies, usually to very recently exposed individuals. At about the same time such approaches were also tried to treat a variety of cancers - proof of principle for the protective role of the immune response against malignancy was established by the observation that tumors transplanted into syngeneic hosts were rejected by the host innate and adaptive responses. The impact of these therapeutic vaccination has taken a considerable time to become established - in part because target antigens against which an adaptive response can be directed do not appear to be uniquely expressed on malignant transformed cells; and also because tumor cells are able to manipulate their environment to downregulate the host immune response. Therapeutic cancer vaccines are also divided into active and passive types - the latter being subdivided into specific and non-specific vaccines. Active immunization utilizes an immunogen to generate a host response designed to eliminate the malignant cells, whereas in passive immunization preformed antibodies or cells are administered to directly eliminate the transformed cells - examples of each are considered in this review.
Collapse
Affiliation(s)
- David Baxter
- a Honorary Lecturer; Manchester University Medical School; Manchester, UK
| |
Collapse
|
157
|
Menderes G, Schwab CL, Black J, Santin AD. The Role of the Immune System in Ovarian Cancer and Implications on Therapy. Expert Rev Clin Immunol 2016; 12:681-95. [PMID: 26821930 DOI: 10.1586/1744666x.2016.1147957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ovarian cancer is the leading cause of death from gynecologic malignancy in the United States. While the treatment options have improved with conventional cytotoxic chemotherapy and advanced surgical techniques, disease recurrence is common and fatal in nearly all cases. Current evidence suggests that the immune system and its ability to recognize and eliminate microscopic disease is paramount in preventing recurrence. The goal of immunotherapy is to balance the activation of the immune system against cancer while preventing the potential for tremendous toxicity elicited by immune modulation. In this paper we will review the role of immune system in disease pathogenesis and different immunotherapies available for the treatment of ovarian cancer as well as current ongoing studies and potential future directions.
Collapse
Affiliation(s)
- Gulden Menderes
- a Department of Obstetrics, Gynecology & Reproductive Sciences , Yale University School of Medicine , New Haven , CT , USA
| | - Carlton L Schwab
- a Department of Obstetrics, Gynecology & Reproductive Sciences , Yale University School of Medicine , New Haven , CT , USA
| | - Jonathan Black
- a Department of Obstetrics, Gynecology & Reproductive Sciences , Yale University School of Medicine , New Haven , CT , USA
| | - Alessandro D Santin
- a Department of Obstetrics, Gynecology & Reproductive Sciences , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
158
|
Holle AW, Young JL, Spatz JP. In vitro cancer cell-ECM interactions inform in vivo cancer treatment. Adv Drug Deliv Rev 2016; 97:270-9. [PMID: 26485156 DOI: 10.1016/j.addr.2015.10.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/05/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023]
Abstract
The general progression of cancer drug development involves in vitro testing followed by safety and efficacy evaluation in clinical trials. Due to the expense of bringing candidate drugs to trials, in vitro models of cancer cells and tumor biology are required to screen drugs. There are many examples of drugs exhibiting cytotoxic behavior in cancer cells in vitro but losing efficacy in vivo, and in many cases, this is the result of poorly understood chemoresistant effects conferred by the cancer microenvironment. To address this, improved methods for culturing cancer cells in biomimetic scaffolds have been developed; along the way, a great deal about the nature of cancer cell-extracellular matrix (ECM) interactions has been discovered. These discoveries will continue to be leveraged both in the development of novel drugs targeting these interactions and in the fabrication of biomimetic substrates for efficient cancer drug screening in vitro.
Collapse
|
159
|
Exploiting IL-17-producing CD4+ and CD8+ T cells to improve cancer immunotherapy in the clinic. Cancer Immunol Immunother 2016; 65:247-59. [PMID: 26825102 DOI: 10.1007/s00262-016-1797-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/12/2016] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is one the most effective approaches for treating patients with tumors, as it bolsters the generation and persistence of memory T cells. In preclinical work, it has been reported that adoptively transferred CD4+ and CD8+ lymphocytes that secrete IL-17A (i.e., Th17 and Tc17 cells) regress tumors to a greater extent than IFN-γ(+)Th1 or Tc1 cells in vivo. Herein, we review the mechanisms underlying how infused Th17 and Tc17 cells regress established malignancies in clinically relevant mouse models of cancer. We also discuss how unique signaling cues--such as co-stimulatory molecules (ICOS and 41BB), cytokines (IL-12 and IL-23) or pharmaceutical reagents (Akt inhibitors, etc.)--can be exploited to bolster the therapeutic potential of IL-17(+) lymphocytes with an emphasis on using this knowledge to improve next-generation clinical trials for patients with cancer.
Collapse
|
160
|
Younis RH, Han KL, Webb TJ. Human Head and Neck Squamous Cell Carcinoma-Associated Semaphorin 4D Induces Expansion of Myeloid-Derived Suppressor Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:1419-29. [PMID: 26740106 DOI: 10.4049/jimmunol.1501293] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 11/25/2015] [Indexed: 01/05/2023]
Abstract
One of the mechanisms by which malignancies can induce immune suppression is through the production of cytokines that affect the maturation and differentiation of inflammatory cells in the tumor microenvironment. Semaphorin 4D (Sema4D) is a proangiogenic cytokine produced by several malignancies, which has been described in the regulation of the immune system. In the present study, we examined the role of human head and neck squamous cell carcinoma (HNSCC)-secreted Sema4D on myeloid cell differentiation. CD33(+) cells cultured in HNSCC cell line-derived conditioned medium differentiated into myeloid derived suppressor cells (MDSC) (CD33(+)CD11b(+)HLA-DR(-/low)). The addition of anti-Sema4D Ab to HNSCC conditioned medium significantly reduced the expansion of the MDSC population. Similarly, knockdown of Sema4D in an HNSCC cell line resulted in a loss of MDSC function as shown by a decrease in the production of the immune-suppressive cytokines arginase-1, TGF-β, and IL-10 by MDSC, concomitant with recovery of T cell proliferation and IFN-γ production following stimulation of CD3/CD28. Importantly, CD33(+) myeloid and T cells cultured in conditioned medium of HNSCC cells in which Sema4D was knocked down promoted antitumor inflammatory profile, through recovery of the effector T cells (CD4(+)T-bet(+) and CD8(+)T-bet(+)), as well as a decrease in regulatory T cells (CD4(+)CD25(+)FOXP3(+)). We also showed that Sema4D was comparable to GM-CSF in its induction of MDSC. Collectively, this study describes a novel immunosuppressive role for Sema4D in HNSCC through induction of MDSC, and it highlights Sema4D as a therapeutic target for future studies to enhance the antitumorigenic inflammatory response in HNSCC and other epithelial malignancies.
Collapse
Affiliation(s)
- Rania H Younis
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD 21201; Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201; and
| | - Kyu Lee Han
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD 21201
| | - Tonya J Webb
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201; and Department of Microbiology and Immunology, University of Maryland, Baltimore, Baltimore, MD 21201
| |
Collapse
|
161
|
Epstein-Barr virus infection and nasopharyngeal carcinoma: the other side of the coin. Anticancer Drugs 2015; 26:1017-25. [PMID: 26241803 DOI: 10.1097/cad.0000000000000276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oncogenic viruses may have a significant impact on the therapeutic management of several malignancies besides their well-known role in tumor pathogenesis. Epstein-Barr virus (EBV) induces neoplastic transformation of epithelial cells of the nasopharynx by various molecular mechanisms mostly involving activation of oncogenes and inactivation of tumor-suppressor genes. EBV infection can also induce the expression of several immunogenic peptides on the plasma membrane of the infected cells. Importantly, these virus-related antigens may be used as targets for antitumor immunotherapy-based treatment strategies. Two different immunotherapy strategies, namely adoptive and active immunotherapy, have been developed and strongly improved in the recent years. Furthermore, EBV infection may influence the use of targeted therapies for nasopharyngeal carcinoma (NPC) considering that the presence of EBV can induce important modifications in cell signaling. As an example, latent membrane protein type 1 is a viral transmembrane protein mainly involved in the cancerogenesis process, which can also mediate overexpression of the epidermal growth factor receptor (EGFR) in NPC cells, rendering them more sensitive to anti-EGFR therapy. Finally, EBV may induce epigenetic changes in the infected cells, such as DNA hypermethylation and histone deacetylation, that can sustain tumor growth and can thus be considered potential targets for novel therapies. In conclusion, EBV infection can modify important biological features of NPC cells, rendering them more vulnerable to both immunotherapy and targeted therapy.
Collapse
|
162
|
Peng Z, Liang W, Li Z, Xu Y, Chen L. Interleukin-15-transferred cytokine-induced killer cells elevated anti-tumor activity in a gastric tumor-bearing nude mice model. Cell Biol Int 2015; 40:204-13. [PMID: 26503216 DOI: 10.1002/cbin.10553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/23/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Zheng Peng
- Department of General Surgery; Chinese PLA General Hospital; Beijing 100853 China
| | - Wentao Liang
- Institute of General Surgery; Chinese PLA General Hospital; Beijing 100853 China
| | - Zexue Li
- Institute of General Surgery; Chinese PLA General Hospital; Beijing 100853 China
| | - Yingxin Xu
- Institute of General Surgery; Chinese PLA General Hospital; Beijing 100853 China
| | - Lin Chen
- Department of General Surgery; Chinese PLA General Hospital; Beijing 100853 China
| |
Collapse
|
163
|
Fang RH, Kroll AV, Zhang L. Nanoparticle-Based Manipulation of Antigen-Presenting Cells for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5483-96. [PMID: 26331993 PMCID: PMC4641138 DOI: 10.1002/smll.201501284] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/20/2015] [Indexed: 05/18/2023]
Abstract
Immunotherapeutic approaches for treating cancer overall have been receiving a considerable amount of interest due to the recent approval of several clinical formulations. Among the different modalities, anticancer vaccination acts by training the body to endogenously generate a response against tumor cells. However, despite the large amount of work that has gone into the development of such vaccines, the near absence of clinically approved formulations highlights the many challenges facing those working in the field. The generation of potent endogenous anticancer responses poses unique challenges due to the similarity between cancer cells and normal, healthy cells. As researchers continue to tackle the limited efficacy of vaccine formulations, fresh and novel approaches are being sought after to address many of the underlying problems. Here the application of nanoparticle technology towards the development of anticancer vaccines is discussed. Specifically, there is a focus on the benefits of using such strategies to manipulate antigen presenting cells (APCs), which are essential to the vaccination process, and how nanoparticle-based platforms can be rationally engineered to elicit appropriate downstream immune responses.
Collapse
Affiliation(s)
- Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashley V. Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
164
|
Chan IH, Wu V, McCauley S, Grimm EA, Mumm JB. IL-10: Expanding the Immune Oncology Horizon. RECEPTORS & CLINICAL INVESTIGATION 2015; 2:1041. [PMID: 26661378 PMCID: PMC4675350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Recent advances in immunoncology have dramatically changed the treatment options available to cancer patients. However, the fundamental challenges with this therapeutic modality are not new and still persist with the current wave of immunoncology compounds. These challenges are centered on the activation and expansion, induction of intratumoral infiltration and persistence of highly activated, cytotoxic, tumor antigen specific CD8+ T cells. We have investigated the anti-tumor mechanism of action of pegylated recombinant interleukin-10, (PEG-rIL-10) both pre-clinically with murine (PEG-rMuIL-10) and now clinically (AM0010) with human pegylated interleukin-10. The preponderance of data suggest that IL-10's engagement of its receptor on CD8+ T cells enhances their activation status leading to antigen specific expansion. Quantitation of CD8+ T cell tumor infiltration reveals that treatment of both humans and mice with pegylated rIL-10 results in 3-4 fold increases of intratumoral, cytotoxic, CD8+ T cells. In addition, mice cured of their tumors with PEG-rMuIL-10 exhibit long term immunological protection from tumor re-challenge and long term treatment of cancer patients with AM0010 results in the persistence of highly activated CD8+ T cells. Cumulatively, these data suggest the IL-10 represents an emerging therapeutic that specifically addresses the fundamental challenges of the current wave of immunoncology assets.
Collapse
Affiliation(s)
- Ivan H. Chan
- ARMO BioSciences 575 Chesapeake Drive Redwood City, CA 94063
| | - Victoria Wu
- ARMO BioSciences 575 Chesapeake Drive Redwood City, CA 94063
| | - Scott McCauley
- ARMO BioSciences 575 Chesapeake Drive Redwood City, CA 94063
| | - Elizabeth A. Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Box 421, 1515 Holcombe Blvd., Houston, TX 77030
| | - John B. Mumm
- ARMO BioSciences 575 Chesapeake Drive Redwood City, CA 94063
| |
Collapse
|
165
|
Rapp M, Grassmann S, Chaloupka M, Layritz P, Kruger S, Ormanns S, Rataj F, Janssen KP, Endres S, Anz D, Kobold S. C-C chemokine receptor type-4 transduction of T cells enhances interaction with dendritic cells, tumor infiltration and therapeutic efficacy of adoptive T cell transfer. Oncoimmunology 2015; 5:e1105428. [PMID: 27195186 PMCID: PMC4859768 DOI: 10.1080/2162402x.2015.1105428] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/02/2015] [Accepted: 10/03/2015] [Indexed: 12/11/2022] Open
Abstract
T cell infiltration at the tumor site has been identified as a major predictor for the efficacy of adoptive T cell therapy. The chemokine C-C motif ligand 22 (CCL22) is highly expressed by immune cells in murine and human pancreatic cancer. Expression of its corresponding receptor, C-C chemokine receptor type 4 (CCR4), is restricted to regulatory T cells (Treg). We show that transduction of cytotoxic T cells (CTL) with CCR4 enhances their immigration into a pancreatic cancer model. Further, we show that binding of CCR4 with CCL22 strengthens the binding of T cell LFA-1 to dendritic cell (DC) ICAM-1 and increases CTL activation. In vivo, in a model of subcutaneous pancreatic cancer, treatment of tumor-bearing mice with CCR4-transduced CTL led to the eradication of established tumors in 40% of the mice. In conclusion, CCR4 overexpression in CTL is a promising therapeutic strategy to enhance the efficacy of adoptive T cell transfer (ACT).
Collapse
Affiliation(s)
- Moritz Rapp
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - Simon Grassmann
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - Michael Chaloupka
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - Patrick Layritz
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - Stephan Kruger
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum der Ludwig-Maximilians-Universität München , Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-Universität München , Munich, Germany
| | - Felicitas Rataj
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München , Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research, Munich, Germany; Department of Internal Medicine IV, Division of Gastroenterology, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research , Munich, Germany
| |
Collapse
|
166
|
In-vitro Optimization of Nanoparticle-Cell Labeling Protocols for In-vivo Cell Tracking Applications. Sci Rep 2015; 5:15400. [PMID: 26507853 PMCID: PMC4623670 DOI: 10.1038/srep15400] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/22/2015] [Indexed: 12/19/2022] Open
Abstract
Recent advances in theranostic nanomedicine can promote stem cell and immune cell-based therapy. Gold nanoparticles (GNPs) have been shown to be promising agents for in-vivo cell-tracking in cell-based therapy applications. Yet a crucial challenge is to develop a reliable protocol for cell upload with, on the one hand, sufficient nanoparticles to achieve maximum visibility of cells, while on the other hand, assuring minimal effect of particles on cell function and viability. Previous studies have demonstrated that the physicochemical parameters of GNPs have a critical impact on their efficient uptake by cells. In the current study we have examined possible variations in GNP uptake, resulting from different incubation period and concentrations in different cell-lines. We have found that GNPs effectively labeled three different cell-lines - stem, immune and cancer cells, with minimal impairment to cell viability and functionality. We further found that uptake efficiency of GNPs into cells stabilized after a short period of time, while GNP concentration had a significant impact on cellular uptake, revealing cell-dependent differences. Our results suggest that while heeding the slight variations within cell lines, modifying the loading time and concentration of GNPs, can promote cell visibility in various nanoparticle-dependent in-vivo cell tracking and imaging applications.
Collapse
|
167
|
Abstract
The therapeutic potential of dendritic cell (DC) cancer vaccines has gained momentum in recent years. However, clinical data indicate that antitumor immune responses generally fail to translate into measurable tumor regression. This has been ascribed to a variety of tolerance mechanisms, one of which is the expression of immunosuppressive factors by DCs and T cells. With respect to cancer immunotherapies, these factors antagonise the ability to induce robust and sustained immunity required for tumor cell eradication. Gene silencing of immunosuppressive factors in either DCs or adoptive transferred T cells enhanced anti-tumor immune responses and significantly inhibited tumor growth. Therefore, engineered next generation of DC vaccines or adoptive T-cell therapy should include immunomodulatory siRNAs to release the "brakes" imposed by the immune system. Moreover, the combination of gene silencing, antigen targeting to DCs and cytoplasmic cargo delivery will improve clinical benefits.
Collapse
Key Words
- AML, acute myeloid leukemia
- CMV, human cytomegalovirus
- CTLA4, T-lymphocyte-associated antigen 4
- DC, Dendritic cells
- Gal, galectin hTERT, human telomerase reverse transcriptase
- IDO, indoleamine 2,3-dioxygenase
- IL, interleukin
- INF, interferon
- NK, natural killer
- PD1, programmed cell death
- RNA interference
- RNAi, RNA interference
- SOCS1, suppressor of cytokine signaling
- STAT, Signal transducer and activator of transcription
- T-cell therapy
- TCR, T cell receptor
- TLR, toll like receptor
- Treg, Regulatory T
- cancer vaccine
- gene silencing
- immunotherapy
- siRNA, small interfering RNA
- targeted therapies
Collapse
Affiliation(s)
- Mouldy Sioud
- a Department of Immunology; Institute for Cancer Research ; Oslo University Hospital ; Montebello , Norway
| |
Collapse
|
168
|
Shi H, Guo J, Li C, Wang Z. A current review of folate receptor alpha as a potential tumor target in non-small-cell lung cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4989-96. [PMID: 26357465 PMCID: PMC4560517 DOI: 10.2147/dddt.s90670] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lung cancer remains the leading common cause of cancer-related death, with non-small-cell lung cancer (NSCLC) accounting for 80% of all cases. To date, platinum-based doublet chemotherapy is the cornerstone of first-line therapy. However, these agents have limited use in patients who have relapsed and have metastatic disease. Therefore, novel strategies are required to improve the clinical outcome. Folate receptor alpha (FRA) is overexpressed in the majority of NSCLC, particularly in lung adenocarcinomas. FRA is largely absent from normal tissue, making it an attractive therapeutic target. In this review, we discuss FRA expression in NSCLC, conjugated FRA agents, monoclonal antibody, and FRA-specific T-cell-based therapeutic strategies aiming to improve the cure rate of FRA-expressing NSCLC.
Collapse
Affiliation(s)
- Huan Shi
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Jun Guo
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Changzheng Li
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Zhehai Wang
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| |
Collapse
|
169
|
Abstract
The immune system is designed to discriminate between self and tumor tissue. Through genetic recombination, there is fundamentally no limit to the number of tumor antigens that immune cells can recognize. Yet, tumors use a variety of immunosuppressive mechanisms to evade immunity. Insight into how the immune system interacts with tumors is expanding rapidly and has accelerated the translation of immunotherapies into medical breakthroughs. Herein, we appraise novel strategies that exploit the patient's immune system to kill cancer. We review various forms of immune-based therapies, which have shown significant promise in patients with hematologic malignancies, including (i) conventional monoclonal therapies like rituximab; (ii) engineered monoclonal antibodies called bispecific T-cell engagers; (iii) monoclonal antibodies and pharmaceutical drugs that block inhibitory T-cell pathways (i.e. PD-1, CTLA-4, and IDO); and (iv) adoptive cell transfer therapy with T cells engineered to express chimeric antigen receptors or T-cell receptors. We also assess the idea of using these therapies in combination and conclude by suggesting multi-prong approaches to improve treatment outcomes and curative responses in patients.
Collapse
Affiliation(s)
- Michelle H Nelson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA; Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
170
|
Anguille S, Smits EL, Bryant C, Van Acker HH, Goossens H, Lion E, Fromm PD, Hart DN, Van Tendeloo VF, Berneman ZN. Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy. Pharmacol Rev 2015; 67:731-53. [DOI: 10.1124/pr.114.009456] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
171
|
Nakagawa Y, Negishi Y, Shimizu M, Takahashi M, Ichikawa M, Takahashi H. Effects of extracellular pH and hypoxia on the function and development of antigen-specific cytotoxic T lymphocytes. Immunol Lett 2015. [PMID: 26209187 DOI: 10.1016/j.imlet.2015.07.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The major effector cells for cellular adaptive immunity are CD8(+) cytotoxic T lymphocytes (CTLs), which can recognize and kill virus-infected cells and tumor cells. Although CTLs exhibit strong cytolytic activity against target cells in vitro, a number of studies have demonstrated that their function is often impaired within tumors. Nevertheless, CTLs can regain their cytotoxic ability after escaping from the tumor environment, suggesting that the milieu created by tumors may affect the function of CTLs. As for the tumor environment, the patho-physiological situation present in vivo has been shown to differ from in vitro experimental conditions. In particular, low pH and hypoxia are the most important microenvironmental factors within growing tumors. In the present study, to determine the effect of these factors on CTL function in vivo, we examined the cytolytic activity of CTLs against their targets using murine CTL lines and the induction of these cells from memory cells under low pH or hypoxic conditions using antigen-primed spleen cells. The results indicated that both cytotoxic activity and the induction of functional CTLs were markedly inhibited under low pH. In contrast, in hypoxic conditions, although cytotoxic activity was almost unchanged, the induction of CTLs in vitro showed a slight enhancement, which was completely abrogated in low pH conditions. Therefore, antigen-specific CTL functions may be more vulnerable to low pH than to the oxygen concentration in vivo. The findings shown here provide new therapeutic approaches for controlling tumor growth by retaining CTL cytotoxicity through the maintenance of higher pH conditions.
Collapse
Affiliation(s)
- Yohko Nakagawa
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan.
| | - Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Megumi Takahashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Masao Ichikawa
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Hidemi Takahashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
172
|
Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R, Lebuhotel C, Eyquem J, Cheung GWK, Duclert A, Gouble A, Arnould S, Peggs K, Pule M, Scharenberg AM, Smith J. Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies. Cancer Res 2015; 75:3853-64. [PMID: 26183927 DOI: 10.1158/0008-5472.can-14-3321] [Citation(s) in RCA: 434] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/10/2015] [Indexed: 12/28/2022]
Abstract
Adoptive immunotherapy using autologous T cells endowed with chimeric antigen receptors (CAR) has emerged as a powerful means of treating cancer. However, a limitation of this approach is that autologous CAR T cells must be generated on a custom-made basis. Here we show that electroporation of transcription activator-like effector nuclease (TALEN) mRNA allows highly efficient multiplex gene editing in primary human T cells. We use this TALEN-mediated editing approach to develop a process for the large-scale manufacturing of T cells deficient in expression of both their αβ T-cell receptor (TCR) and CD52, a protein targeted by alemtuzumab, a chemotherapeutic agent. Functionally, T cells manufactured with this process do not mediate graft-versus-host reactions and are rendered resistant to destruction by alemtuzumab. These characteristics enable the administration of alemtuzumab concurrently or prior to engineered T cells, supporting their engraftment. Furthermore, endowing the TALEN-engineered cells with a CD19 CAR led to efficient destruction of CD19(+) tumor targets even in the presence of the chemotherapeutic agent. These results demonstrate the applicability of TALEN-mediated genome editing to a scalable process, which enables the manufacturing of third-party CAR T-cell immunotherapies against arbitrary targets. As such, CAR T-cell immunotherapies can therefore be used in an "off-the-shelf" manner akin to other biologic immunopharmaceuticals
Collapse
Affiliation(s)
| | - Brian Philip
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | - Gordon Weng-Kit Cheung
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | | | | | | | - Karl Peggs
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Martin Pule
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Andrew M Scharenberg
- Department of Pediatrics, University of Washington, Seattle Children's Research Institute, Seattle, Washington
| | | |
Collapse
|
173
|
Sampath P, Thorne SH. Novel therapeutic strategies in human malignancy: combining immunotherapy and oncolytic virotherapy. Oncolytic Virother 2015; 4:75-82. [PMID: 27512672 PMCID: PMC4918382 DOI: 10.2147/ov.s54738] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Results from randomized clinical trials over the last several years have finally begun to demonstrate the potential of oncolytic viral therapies to treat a variety of cancers. One reason for these successes has been the realization that this platform is most effective when considered primarily as an immunotherapy. Cancer immunotherapy has also made dramatic strides recently with antibodies capable of blocking immune checkpoint inhibitors and adoptive T-cell therapies, notably CAR T-cells, leading a panel of novel and highly clinically effective therapies. It is clear therefore that an understanding of how and when these complementary approaches can most effectively be combined offers the real hope of moving beyond simply treating the disease and toward starting to talk about curative therapies. In this review we discuss approaches to combining these therapeutic platforms, both through engineering the viral vectors to more beneficially interact with the host immune response during therapy, as well as through the direct combinations of different therapeutics. This primarily, but not exclusively focuses on strains of oncolytic vaccinia virus. Some of the results reported to date, primarily in pre-clinical models but also in early clinical trials, are dramatic and hold great promise for the future development of similar therapies and their translation into cancer therapies.
Collapse
Affiliation(s)
- Padma Sampath
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steve H Thorne
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
174
|
Robertson-Tessi M, El-Kareh A, Goriely A. A model for effects of adaptive immunity on tumor response to chemotherapy and chemoimmunotherapy. J Theor Biol 2015; 380:569-84. [PMID: 26087282 DOI: 10.1016/j.jtbi.2015.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 05/08/2015] [Accepted: 06/02/2015] [Indexed: 10/23/2022]
Abstract
Complete clinical regressions of solid tumors in response to chemotherapy are difficult to explain by direct cytotoxicity alone, because of low growth fractions and obstacles to drug delivery. A plausible indirect mechanism that might reconcile this is the action of the immune system. A model for interaction between tumors and the adaptive immune system is presented here, and used to examine controllability of tumors through the interplay of cytotoxic, cytostatic and immunogenic effects of chemotherapy and the adaptive immune response. The model includes cytotoxic and helper T cells, T regulatory cells (Tregs), dendritic cells, memory cells, and several key cytokines. Nearly all parameter estimates are derived from experimental and clinical data. Individual tumors are characterized by two parameters: growth rate and antigenicity, and regions of tumor control are identified in this parameter space. The model predicts that inclusion of the immune response significantly expands the region of tumor control for both cytostatic and cytotoxic chemotherapies. Moreover, outside the control zone, tumor growth is delayed significantly. An optimal fractionation schedule is predicted, for a fixed cumulative dose. The model further predicts expanded regions of tumor control when several forms of immunotherapy (adoptive T cell transfer, Treg depletion, and dendritic cell vaccination) are combined with chemotherapy. Outcomes depend greatly on tumor characteristics, the schedule of administration, and the type of immunotherapy chosen, suggesting promising opportunities for personalized medicine. Overall, the model provides insight into the role of the adaptive immune system in chemotherapy, and how scheduling and immunotherapeutic interventions might improve efficacy.
Collapse
Affiliation(s)
- Mark Robertson-Tessi
- Program in Applied Mathematics, University of Arizona, Tucson, AZ 85721, United States; Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL 33612, United States.
| | - Ardith El-Kareh
- ARL-Microcirculation Division, University of Arizona, Tucson, AZ 85724, United States
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Woodstock Road, OX2 6GG, UK
| |
Collapse
|
175
|
Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving both naturally occurring tumor immunity and immunotherapy. Nat Immunol 2015; 16:850-8. [PMID: 26075911 DOI: 10.1038/ni.3201] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/17/2015] [Indexed: 12/12/2022]
Abstract
The success of antitumor immune responses depends on the infiltration of solid tumors by effector T cells, a process guided by chemokines. Here we show that in vivo post-translational processing of chemokines by dipeptidylpeptidase 4 (DPP4, also known as CD26) limits lymphocyte migration to sites of inflammation and tumors. Inhibition of DPP4 enzymatic activity enhanced tumor rejection by preserving biologically active CXCL10 and increasing trafficking into the tumor by lymphocytes expressing the counter-receptor CXCR3. Furthermore, DPP4 inhibition improved adjuvant-based immunotherapy, adoptive T cell transfer and checkpoint blockade. These findings provide direct in vivo evidence for control of lymphocyte trafficking via CXCL10 cleavage and support the use of DPP4 inhibitors for stabilizing biologically active forms of chemokines as a strategy to enhance tumor immunotherapy.
Collapse
|
176
|
Haji-Fatahaliha M, Hosseini M, Akbarian A, Sadreddini S, Jadidi-Niaragh F, Yousefi M. CAR-modified T-cell therapy for cancer: an updated review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1339-49. [PMID: 26068778 DOI: 10.3109/21691401.2015.1052465] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The use of chimeric antigen receptor (CAR)-modified T cells is a promising approach for cancer immunotherapy. These genetically modified receptors contain an antigen-binding moiety, a hinge region, a transmembrane domain, and an intracellular costimulatory domain resulting in T-cell activation subsequent to antigen binding. Optimal tumor removal through CAR-modified T cells requires suitable target antigen selection, co-stimulatory signaling domain, and the ability of CAR T cells to traffic, persist, and retain antitumor function after adoptive transfer. There are several elements which can improve antitumor function of CAR T cells, including signaling, conditioning chemotherapy and irradiation, tumor burden of the disease, T-cell phenotype, and supplementary cytokine usage. This review outlines four generations of CAR. The pre-clinical and clinical studies showed that this technique has a great potential for treatment of solid and hematological malignancies. The main purpose of the current review is to focus on the pre-clinical and clinical developments of CAR-based immunotherapy.
Collapse
Affiliation(s)
- Mostafa Haji-Fatahaliha
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Maryam Hosseini
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Asiye Akbarian
- d Department of Microbiology , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Sanam Sadreddini
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Farhad Jadidi-Niaragh
- e Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
177
|
Yue C, Shen S, Deng J, Priceman SJ, Li W, Huang A, Yu H. STAT3 in CD8+ T Cells Inhibits Their Tumor Accumulation by Downregulating CXCR3/CXCL10 Axis. Cancer Immunol Res 2015; 3:864-870. [PMID: 26025380 DOI: 10.1158/2326-6066.cir-15-0014] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/20/2015] [Indexed: 12/11/2022]
Abstract
One of the obstacles for cancer immunotherapy is the inefficiency of CD8(+) T-cell recruitment to tumors. STAT3 has been shown to suppress CD8(+) T-cell antitumor functions in various cancer models, in part by restricting accumulation of CD8(+) T cells. However, the underlying molecular mechanism by which STAT3 in CD8(+) T cells inhibits their accumulation in tumors remains to be defined. Here, we show that STAT3 signaling in CD8(+) T cells inhibits chemokine CXCL10 production by tumor-associated myeloid cells by reducing IFNγ expression by T cells. We further demonstrate that ablating STAT3 in T cells allows expression of CXCR3, the receptor of CXCL10, on CD8(+) T cells, resulting in efficient accumulation of CD8(+) T cells at tumor sites. Blocking IFNγ or CXCR3 impairs the accumulation of STAT3-deficient CD8(+) T cells in tumor and their antitumor effects. Together, our study reveals a negative regulation by STAT3 signaling in T cells on cross-talk between myeloid cells and T cells through IFNγ/CXCR3/CXCL10, which is important for CD8(+) T cells homing to tumors. Our results thus provide new insights applicable to cancer immunotherapy and adoptive T-cell strategies.
Collapse
Affiliation(s)
- Chanyu Yue
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Shudan Shen
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jiehui Deng
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Saul J Priceman
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Wenzhao Li
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Austin Huang
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Hua Yu
- Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
178
|
Greco R, Oliveira G, Stanghellini MTL, Vago L, Bondanza A, Peccatori J, Cieri N, Marktel S, Mastaglio S, Bordignon C, Bonini C, Ciceri F. Improving the safety of cell therapy with the TK-suicide gene. Front Pharmacol 2015; 6:95. [PMID: 25999859 PMCID: PMC4419602 DOI: 10.3389/fphar.2015.00095] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/17/2015] [Indexed: 01/07/2023] Open
Abstract
While opening new frontiers for the cure of malignant and non-malignant diseases, the increasing use of cell therapy poses also several new challenges related to the safety of a living drug. The most effective and consolidated cell therapy approach is allogeneic hematopoietic stem cell transplantation (HSCT), the only cure for several patients with high-risk hematological malignancies. The potential of allogeneic HSCT is strictly dependent on the donor immune system, particularly on alloreactive T lymphocytes, that promote the beneficial graft-versus-tumor effect (GvT), but may also trigger the detrimental graft-versus-host-disease (GvHD). Gene transfer technologies allow to manipulate donor T-cells to enforce GvT and foster immune reconstitution, while avoiding or controlling GvHD. The suicide gene approach is based on the transfer of a suicide gene into donor lymphocytes, for a safe infusion of a wide T-cell repertoire, that might be selectively controlled in vivo in case of GvHD. The herpes simplex virus thymidine kinase (HSV-TK) is the suicide gene most extensively tested in humans. Expression of HSV-TK in donor lymphocytes confers lethal sensitivity to the anti-herpes drug, ganciclovir. Progressive improvements in suicide genes, vector technology and transduction protocols have allowed to overcome the toxicity of GvHD while preserving the antitumor efficacy of allogeneic HSCT. Several phase I-II clinical trials in the last 20 years document the safety and the efficacy of HSV-TK approach, able to maintain its clear value over the last decades, in the rapidly progressing horizon of cancer cellular therapy.
Collapse
Affiliation(s)
- Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Giacomo Oliveira
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Program in Immunology and Bio-immunotherapy of Cancer, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Maria Teresa Lupo Stanghellini
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Luca Vago
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy ; Unit of Molecular and Functional Immunogenetics, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Attilio Bondanza
- Leukemia Immunotherapy Unit, Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Jacopo Peccatori
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Nicoletta Cieri
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Program in Immunology and Bio-immunotherapy of Cancer, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Sarah Marktel
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Sara Mastaglio
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Program in Immunology and Bio-immunotherapy of Cancer, IRCCS San Raffaele Scientific Institute, Milan Italy
| | | | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Program in Immunology and Bio-immunotherapy of Cancer, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan Italy
| |
Collapse
|
179
|
Karlitepe A, Ozalp O, Avci CB. New approaches for cancer immunotherapy. Tumour Biol 2015; 36:4075-8. [PMID: 25934338 DOI: 10.1007/s13277-015-3491-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/23/2015] [Indexed: 11/28/2022] Open
Abstract
Immunotherapy is a promising field that offers alternative methods for treatment of cancer. The current strategy consists of cancer vaccines, monoclonal antibodies, and cellular therapies. Cancer vaccines aim to eradicate cancer cells via immune system. Thus, they may attack these cells derived from any type of cancer, besides their role in preventing cancer. Lymphocytes and dendritic cells are often used in cellular therapy. In addition, monoclonal antibodies are designed to target specific antigens found in cancer cells. Currently, at least 12 clinically approved monoclonal antibodies are being used and many cancer vaccines are being developed with ongoing phase studies for cancer therapy. Relevant studies are focused on glioma and several other cancer types. Correspondingly, the combination of effective methods may enhance the efficacy of immunotherapy. It is thought that particularly immune checkpoint inhibitors will play a crucial role in immunotherapeutic approaches.
Collapse
Affiliation(s)
- Ayfer Karlitepe
- Department of Medical Biochemistry, Medical School, Ege University, Izmir, Turkey
| | | | | |
Collapse
|
180
|
Abstract
There is a clear need to develop strategies to induce tolerance without the need of chronic immunosuppression in transplant recipient and in patients with autoimmunity. Adoptive T regulatory cell (Treg) therapy offers the potential of long-lasting protection. However, based on results of clinical trials so far with ex vivo expanded autologous Tregs in type 1 diabetic (T1D) patients, it seems unlikely that single immunotherapy with Treg infusion without immunomodulation regimens that promote stable donor Treg engraftment and persistence would afford truly significant clinical benefit. Combination therapies could provide improved outcomes with consideration of the fundamental factors required for Treg generation, homeostasis, and function to promote long-term donor Treg persistence to provoke beneficial therapeutic outcomes.
Collapse
|
181
|
Abstract
PURPOSE OF REVIEW Epithelial ovarian cancer is the most frequent cause of gynecologic cancer-related mortality in women, and prognosis for patients with recurrent or metastatic disease is extremely poor. Therefore, there is an enormous unmet need for the development of novel therapies in this indication. Although surgery and chemotherapy can improve survival rates, it is necessary to integrate alternative strategies, such as immunotherapy to improve the outcomes for patients with advanced ovarian cancer. RECENT FINDINGS We will discuss the rationale of immunotherapy and some of the mechanisms of immunogenicity in ovarian cancer. We will highlight current results with cancer vaccines, adoptive T-cell therapy and immunomodulatory agents and will summarize the immune effects of selected chemotherapeutic agents, radiotherapy and recent results with combinatorial approaches in this disease setting. We will also discuss recent and potential future therapeutic interventions that might circumvent tumor-mediated immunosuppression. SUMMARY Dramatic increase in the number of immunotherapy clinical trials was seen in the past decade with promising results in enhancing antitumor immune response and cancer vaccine efficacy. The future challenge for immunotherapy against ovarian cancer is to use a combinatorial approach to test rational, potentially synergistic immunotherapy combinations that can induce efficient antitumor immunity and prolong patients' survival.
Collapse
|
182
|
Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol 2015; 35 Suppl:S185-S198. [PMID: 25818339 DOI: 10.1016/j.semcancer.2015.03.004] [Citation(s) in RCA: 1013] [Impact Index Per Article: 112.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/27/2022]
Abstract
Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.
Collapse
|
183
|
Nayar S, Dasgupta P, Galustian C. Extending the lifespan and efficacies of immune cells used in adoptive transfer for cancer immunotherapies-A review. Oncoimmunology 2015; 4:e1002720. [PMID: 26155387 DOI: 10.1080/2162402x.2014.1002720] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/19/2014] [Accepted: 12/20/2014] [Indexed: 12/19/2022] Open
Abstract
Cells used in adoptive cell-transfer immunotherapies against cancer include dendritic cells (DCs), natural-killer cells, and CD8+ T-cells. These cells may have limited efficacy due to their lifespan, activity, and immunosuppressive effects of tumor cells. Therefore, increasing longevity and activity of these cells may boost their efficacy. Four cytokines that can extend immune effector-cell longevity are IL-2, IL-7, IL-21, and IL-15. This review will discuss current knowledge on effector-cell lifespans and the mechanisms by which IL-2, IL-7, IL-15, and IL-21 can extend effector-cell longevity. We will also discuss how lifespan and efficacy of these cells can be regulated to allow optimal clinical benefits.
Collapse
Affiliation(s)
- Sandeep Nayar
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Prokar Dasgupta
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Christine Galustian
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| |
Collapse
|
184
|
Obenaus M, Leitão C, Leisegang M, Chen X, Gavvovidis I, van der Bruggen P, Uckert W, Schendel DJ, Blankenstein T. Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat Biotechnol 2015; 33:402-7. [PMID: 25774714 DOI: 10.1038/nbt.3147] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/12/2015] [Indexed: 12/21/2022]
Abstract
Identifying T-cell receptors (TCRs) that bind tumor-associated antigens (TAAs) with optimal affinity is a key bottleneck in the development of adoptive T-cell therapy of cancer. TAAs are unmutated self proteins, and T cells bearing high-affinity TCRs specific for such antigens are commonly deleted in the thymus. To identify optimal-affinity TCRs, we generated antigen-negative humanized mice with a diverse human TCR repertoire restricted to the human leukocyte antigen (HLA) A*02:01 (ref. 3). These mice were immunized with human TAAs, for which they are not tolerant, allowing induction of CD8⁺ T cells with optimal-affinity TCRs. We isolate TCRs specific for the cancer/testis (CT) antigen MAGE-A1 (ref. 4) and show that two of them have an anti-tumor effect in vivo. By comparison, human-derived TCRs have lower affinity and do not mediate substantial therapeutic effects. We also identify optimal-affinity TCRs specific for the CT antigen NY-ESO. Our humanized mouse model provides a useful tool for the generation of optimal-affinity TCRs for T-cell therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaojing Chen
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Pierre van der Bruggen
- 1] Ludwig Institute for Cancer Research and WELBIO, Brussels, Belgium. [2] De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Wolfgang Uckert
- 1] Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. [2] Institute of Biology, Humboldt University, Berlin, Germany
| | | | - Thomas Blankenstein
- 1] Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. [2] Institute of Immunology, Charité Campus Buch, Berlin, Germany
| |
Collapse
|
185
|
Zhang GQ, Zhao H, Wu JY, Li JY, Yan X, Wang G, Wu LL, Zhang XG, Shao Y, Wang Y, Jiao SC. Prolonged overall survival in gastric cancer patients after adoptive immunotherapy. World J Gastroenterol 2015; 21:2777-2785. [PMID: 25759549 PMCID: PMC4351231 DOI: 10.3748/wjg.v21.i9.2777] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/04/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess the efficacy of immunotherapy with expanded activated autologous lymphocytes (EAALs) in gastric cancer.
METHODS: An observational study was designed to retrospectively analyze the clinical data of 84 gastric cancer patients, of whom 42 were treated by EAAL immunotherapy plus conventional treatment and another 42 only received conventional treatment (control group). EAALs were obtained by proliferation of peripheral blood mononuclear cells from patients followed by phenotype determination. Clinical data including age, gender, clinical stage, chemotherapeutic regimens, hospitalization, surgical, radiotherapy, and survival data were collected along with EAAL therapy details and side effects. Patients were followed and the relationship between treatment and overall survival (OS) data obtained for the immunotherapy and control groups were compared retrospectively. The safety of EAAL immunotherapy was also evaluated.
RESULTS: After in vitro culture and proliferation, the percentages of CD3+, CD3+CD8+, CD8+CD27+, CD8+CD28+, and CD3+CD16+/CD56+ cells increased remarkably (P < 0.05), while the percentages of CD3+CD4+, CD4+CD25+, and CD3-CD16+/CD56+ (natural killer cells) were overtly decreased (P < 0.05); no significant change was observed in CD4+CD25+CD127- cells (P = 0.448). Interestingly, OS in the immunotherapy group was significantly higher than that in the control group, with 27.0 and 13.9 mo obtained for the two groups, respectively (P = 0.028, HR = 0.573, 95%CI: 0.347-0.945). These findings indicated a 42.7% decrease in the risk of death. In addition, we found that clinical stage and application of EAAL immunotherapy were independent prognostic factors for gastric cancer patients. Indeed, the OS in stage IIIc and IV patients that had received surgery was prolonged after EAAL immunotherapy (P < 0.05). Importantly, in vitro induction and proliferation of EAAL were easy and biologically safe.
CONCLUSION: Overall, EAAL adoptive immunotherapy might prolong the OS in gastric cancer patients.
Collapse
|
186
|
Abstract
Clinical outcomes, such as recurrence-free survival and overall survival, in ovarian cancer are quite variable, independent of common characteristics such as stage, response to therapy, and grade. This disparity in outcomes warrants further exploration and therapeutic targeting into the interaction between the tumor and host. One compelling host characteristic that contributes both to the initiation and progression of ovarian cancer is the immune system. Hundreds of studies have confirmed a prominent role for the immune system in modifying the clinical course of the disease. Recent studies also show that anti-tumor immunity is often negated by immune regulatory cells present in the tumor microenvironment. Regulatory immune cells also directly enhance the pathogenesis through the release of various cytokines and chemokines, which together form an integrated pathological network. Thus, in the future, research into immunotherapy targeting ovarian cancer will probably become increasingly focused on combination approaches that simultaneously augment immunity while preventing local immune suppression. In this article, we summarize important immunological targets that influence ovarian cancer outcome as well as include an update on newer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Keith L Knutson
- Cancer Vaccines and Immune Therapies Program, The Vaccine and Gene Therapy Institute of Florida, 9801 SW Discovery Way, Port St. Lucie, FL, 34949, USA,
| | | | | | | |
Collapse
|
187
|
Page AJ, Cosgrove DC, Herman JM, Pawlik TM. Advances in understanding of colorectal liver metastasis and implications for the clinic. Expert Rev Gastroenterol Hepatol 2015; 9:245-59. [PMID: 25033964 DOI: 10.1586/17474124.2014.940897] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Colorectal cancer is one of the most common cancers in both the USA and Europe. Over the course of diagnosis, treatment and surveillance, up to 50% of these patients will develop metastases to their liver. In the past 20 years alone, there have been multiple advances in the management of these colorectal metastases to the liver. These advances have been made in characterization of these tumors, diagnosis and in treatment, both locally and systemically. Because of this progress, there are subsets of patients with this stage IV disease who are cured of their disease. While significant progress has been made, there still exist limitations in the management of metastatic colorectal cancer to the liver. This review outlines current strategies and highlights recent advances in the management of colorectal liver metastases.
Collapse
Affiliation(s)
- Andrew J Page
- Department of Surgery, Johns Hopkins Hospital, Blalock 688, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
188
|
Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ. Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol 2015; 16:1. [PMID: 25636521 PMCID: PMC4318439 DOI: 10.1186/s12865-014-0064-x] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/23/2014] [Indexed: 02/07/2023] Open
Abstract
Background Adoptive transfer of tumor infiltrating or circulating lymphocytes transduced with tumor antigen receptors has been examined in various clinical trials to treat human cancers. The tumor antigens targeted by transferred lymphocytes affects the efficacy of this therapeutic approach. Because cancer stem cells (CSCs) play an important role in tumor growth and metastasis, we hypothesized that adoptive transfer of T cells targeting a CSC antigen could result in dramatic anti-tumor effects. Results An EpCAM-specific chimeric antigen receptor (CAR) was constructed to transduce human peripheral blood lymphocytes (PBLs) and thereby enable them to target the CSC marker EpCAM. To investigate the therapeutic capabilities of PBLs expressing EpCAM-specific CARs, we used two different tumor models, PC3, the human prostate cancer cell line, which has low expression levels of EpCAM, and PC3M, a highly metastatic clone of PC3 that has high expression levels of EpCAM. We demonstrate that CAR-expressing PBLs can kill PC3M tumor cells in vitro and in vivo. Despite the low expression of EpCAM on PC3 cells, CAR-expressing PBLs significantly inhibited tumor growth and prolonged mouse survival in a PC3 metastasis model, probably by targeting the highly proliferative and metastatic population of cancer cells. Conclusions Our data demonstrate that PBLs expressing with EpCAM-specific CARs have significant anti-tumor activity against prostate cancer. Therefore, the adoptive transfer of T cells targeting EpCAM could have great potential as a cancer treatment.
Collapse
Affiliation(s)
- Zhenling Deng
- Department of Immunology, Cancer Hospital & Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Yanhong Wu
- Department of Immunology, Cancer Hospital & Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Wenbo Ma
- Department of Immunology, Cancer Hospital & Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Shuren Zhang
- Department of Immunology, Cancer Hospital & Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Yu-Qian Zhang
- Department of Immunology, Cancer Hospital & Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100021, China.
| |
Collapse
|
189
|
Yan Y, Li S, Jia T, Du X, Xu Y, Zhao Y, Li L, Liang K, Liang W, Sun H, Li R. Combined therapy with CTL cells and oncolytic adenovirus expressing IL-15-induced enhanced antitumor activity. Tumour Biol 2015; 36:4535-43. [PMID: 25627006 DOI: 10.1007/s13277-015-3098-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/09/2015] [Indexed: 12/14/2022] Open
Abstract
Addition of immunoregulation factor to an oncolytic adenovirus being constructed is a developmental step in tumor gene therapy; however, cytokine IL-15 has not been frequently used as a potential cancer therapy agent. Here, we constructed an E2F-1 promoter oncolytic adenovirus based on type 5 adenovirus, which induces viral replication and proliferation in targeted tumor cells. We inserted the IL-15 gene into the E3 region of the model and found that human IL-15 expressing oncolytic adenovirus (Ad-E2F/IL15) shows a more intense antitumor effect than simple oncolytic viruses (Ad-E2F) do. Precisely because IL-15 can activate natural killer (NK) cells, CD8(+)T cells, and other immune cells, in antitumor therapy, Ad-E2F/IL15 was used in combination with cytotoxic T lymphocytes (CTL) to create a virus that can induce IL-15 gene expression while lysing tumors and stimulating the activity and function of adoptive immune cells. The therapeutic effect of this therapy is clearly stronger than that of a single application of oncolytic viruses or CTL, and hence, it could be a potential new tumor therapy.
Collapse
Affiliation(s)
- Yang Yan
- General Surgery Department, Chinese PLA General Hospital, 28 Fuxing Road, Haidian, Beijing, 100853, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Colapicchioni V, Palchetti S, Pozzi D, Marini ES, Riccioli A, Ziparo E, Papi M, Amenitsch H, Caracciolo G. Killing cancer cells using nanotechnology: novel poly(I:C) loaded liposome–silica hybrid nanoparticles. J Mater Chem B 2015; 3:7408-7416. [DOI: 10.1039/c5tb01383f] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Synthesized core–shell liposome–silica hybrid nanoparticles (LSH NPs), when loaded with the anti-cancer polyinosinic–polycytidylic acid (poly(I:C)), exhibit high anti-tumoral activity in prostate and breast cancer cells.
Collapse
Affiliation(s)
| | - Sara Palchetti
- Department of Molecular Medicine
- ‘Sapienza’ University of Rome
- 00161 Rome
- Italy
| | - Daniela Pozzi
- Department of Molecular Medicine
- ‘Sapienza’ University of Rome
- 00161 Rome
- Italy
| | - Elettra Sara Marini
- Istituto Pasteur-Fondazione Cenci Bolognetti
- Department of Anatomy
- Histology
- Forensic Medicine and Orthopaedics
- Section of Histology and Medical Embryology
| | - Anna Riccioli
- Istituto Pasteur-Fondazione Cenci Bolognetti
- Department of Anatomy
- Histology
- Forensic Medicine and Orthopaedics
- Section of Histology and Medical Embryology
| | - Elio Ziparo
- Istituto Pasteur-Fondazione Cenci Bolognetti
- Department of Anatomy
- Histology
- Forensic Medicine and Orthopaedics
- Section of Histology and Medical Embryology
| | - Massimiliano Papi
- Istituto di Fisica
- Università Cattolica del Sacro Cuore
- 00168 Rome
- Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry
- Graz University of Technology
- 8010 Graz
- Austria
| | - Giulio Caracciolo
- Department of Molecular Medicine
- ‘Sapienza’ University of Rome
- 00161 Rome
- Italy
| |
Collapse
|
191
|
MGN1703, an immunomodulator and toll-like receptor 9 (TLR-9) agonist: from bench to bedside. Crit Rev Oncol Hematol 2014; 94:31-44. [PMID: 25577571 DOI: 10.1016/j.critrevonc.2014.12.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/06/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023] Open
Abstract
The adaptive immune system has been the main focus of immunological strategies in oncology with only more recent approaches targeting innate immunity. Endosomal toll-like receptors (TLR-7, TLR-9) activate innate immune responses by signaling damage-associated molecular patterns (DAMP) from decaying tumor cells. This has led to the development of DNA-based TLR-9 agonists, which induce antitumor activity through innate and subsequent adaptive immune responses. Early clinical trials with CpG-ODN as TLR-9 agonists were associated with unfavorable tolerability and narrow clinical efficacy, leading to failure in pivotal trials. dSLIM, the active ingredient of MGN1703, is a DNA-based, radically different molecular alternative to CpG-ODN, which results in genuine antitumor immunomodulation. Preclinical and clinical studies of MGN1703 have confirmed that this TLR-9 agonist has therapeutic potential in a variety of solid tumors, while long-term treatment with high doses was very well tolerated. A pivotal trial of first-line maintenance treatment with MGN1703 in patients with metastatic colorectal cancer is underway.
Collapse
|
192
|
Deniger DC, Moyes JS, Cooper LJN. Clinical applications of gamma delta T cells with multivalent immunity. Front Immunol 2014; 5:636. [PMID: 25566249 PMCID: PMC4263175 DOI: 10.3389/fimmu.2014.00636] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/28/2014] [Indexed: 01/13/2023] Open
Abstract
γδ T cells hold promise for adoptive immunotherapy because of their reactivity to bacteria, viruses, and tumors. However, these cells represent a small fraction (1–5%) of the peripheral T-cell pool and require activation and propagation to achieve clinical benefit. Aminobisphosphonates specifically expand the Vγ9Vδ2 subset of γδ T cells and have been used in clinical trials of cancer where objective responses were detected. The Vγ9Vδ2 T cell receptor (TCR) heterodimer binds multiple ligands and results in a multivalent attack by a monoclonal T cell population. Alternatively, populations of γδ T cells with oligoclonal or polyclonal TCR repertoire could be infused for broad-range specificity. However, this goal has been restricted by a lack of applicable expansion protocols for non-Vγ9Vδ2 cells. Recent advances using immobilized antigens, agonistic monoclonal antibodies (mAbs), tumor-derived artificial antigen presenting cells (aAPC), or combinations of activating mAbs and aAPC have been successful in expanding gamma delta T cells with oligoclonal or polyclonal TCR repertoires. Immobilized major histocompatibility complex Class-I chain-related A was a stimulus for γδ T cells expressing TCRδ1 isotypes, and plate-bound activating antibodies have expanded Vδ1 and Vδ2 cells ex vivo. Clinically sufficient quantities of TCRδ1, TCRδ2, and TCRδ1negTCRδ2neg have been produced following co-culture on aAPC, and these subsets displayed differences in memory phenotype and reactivity to tumors in vitro and in vivo. Gamma delta T cells are also amenable to genetic modification as evidenced by introduction of αβ TCRs, chimeric antigen receptors, and drug-resistance genes. This represents a promising future for the clinical application of oligoclonal or polyclonal γδ T cells in autologous and allogeneic settings that builds on current trials testing the safety and efficacy of Vγ9Vδ2 T cells.
Collapse
Affiliation(s)
- Drew C Deniger
- Surgery Branch, National Cancer Institute , Bethesda, MD , USA
| | - Judy S Moyes
- Division of Pediatrics, University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Laurence J N Cooper
- Division of Pediatrics, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences, UT MD Anderson Cancer Center , Houston, TX , USA
| |
Collapse
|
193
|
Moogk D, da Silva IP, Ma MW, Friedman EB, de Miera EVS, Darvishian F, Scanlon P, Perez-Garcia A, Pavlick AC, Bhardwaj N, Christos PJ, Osman I, Krogsgaard M. Melanoma expression of matrix metalloproteinase-23 is associated with blunted tumor immunity and poor responses to immunotherapy. J Transl Med 2014; 12:342. [PMID: 25491880 PMCID: PMC4272770 DOI: 10.1186/s12967-014-0342-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/24/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Matrix metalloproteinase-23 (MMP-23) can block the voltage-gated potassium channel Kv1.3, whose function is important for sustained Ca(2+) signaling during T cell activation. MMP-23 may also alter T cell activity and phenotype through cleavage of proteins affecting cytokine and chemokine signaling. We therefore tested the hypothesis that MMP-23 can negatively regulate the anti-tumor T cell response in human melanoma. METHODS We characterized MMP-23 expression in primary melanoma patients who received adjuvant immunotherapy. We examined the association of MMP-23 with the anti-tumor immune response - as assessed by the prevalence of tumor-infiltrating lymphocytes and Foxp3(+) regulatory T cells. Further, we examined the association between MMP-23 expression and response to immunotherapy. Considering also an in trans mechanism, we examined the association of melanoma MMP-23 and melanoma Kv1.3 expression. RESULTS Our data revealed an inverse association between primary melanoma MMP-23 expression and the anti-tumor T cell response, as demonstrated by decreased tumor-infiltrating lymphocytes (TIL) (P = 0.05), in particular brisk TILs (P = 0.04), and a trend towards an increased proportion of immunosuppressive Foxp3(+) regulatory T cells (P = 0.07). High melanoma MMP-23 expression is also associated with recurrence in patients treated with immune biologics (P = 0.037) but not in those treated with vaccines (P = 0.64). Further, high melanoma MMP-23 expression is associated with shorter periods of progression-free survival for patients receiving immune biologics (P = 0.025). On the other hand, there is no relationship between melanoma MMP-23 and melanoma Kv1.3 expression (P = 0.27). CONCLUSIONS Our data support a role for MMP-23 as a potential immunosuppressive target in melanoma, as well as a possible biomarker for informing melanoma immunotherapies.
Collapse
Affiliation(s)
- Duane Moogk
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Department of Pathology, New York University School of Medicine, New York, NY, USA.
| | - Ines Pires da Silva
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA. .,Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal. .,Programme for Advanced Medical Education, Lisbon, Portugal.
| | - Michelle W Ma
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| | - Erica B Friedman
- Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA. .,Department of Surgery, New York University School of Medicine, New York, NY, USA.
| | - Eleazar Vega-Saenz de Miera
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| | - Farbod Darvishian
- Department of Pathology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| | - Patrick Scanlon
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| | - Arianne Perez-Garcia
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Department of Pathology, New York University School of Medicine, New York, NY, USA.
| | - Anna C Pavlick
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA. .,Department of Medicine, New York University School of Medicine, New York, NY, USA.
| | - Nina Bhardwaj
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA. .,Department of Medicine, New York University School of Medicine, New York, NY, USA.
| | - Paul J Christos
- Division of Biostatistics and Epidemiology, Weill Cornell Medical College, New York, NY, USA.
| | - Iman Osman
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| | - Michelle Krogsgaard
- Perlmutter Cancer Center at NYU Langone, New York, NY, USA. .,Department of Pathology, New York University School of Medicine, New York, NY, USA. .,Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
194
|
Vinay DS, Kwon BS. 4-1BB (CD137), an inducible costimulatory receptor, as a specific target for cancer therapy. BMB Rep 2014; 47:122-9. [PMID: 24499671 PMCID: PMC4163883 DOI: 10.5483/bmbrep.2014.47.3.283] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 12/30/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Although considerable progress has been made in understanding how tumors evade immune surveillance, measures to counter the same have not kept pace with the advances made in designing effective strategies. 4-1BB (CD137; TNFRS9), an activation-induced costimulatory molecule, is an important regulator of immune responses. Targeting 4-1BB or its natural ligand 4-1BB ligand (4-1BBL) has important implications in many clinical conditions, including cancer. In-depth analysis revealed that 4-1BB-mediated anti-cancer effects are based on its ability to induce activation of cytotoxic T lymphocytes (CTL), and among others, high amounts of IFN-γ. In this review, we will discuss the various aspects of 4-1BB-mediated anti-tumor responses, the basis of such responses, and future directions. [BMB Reports 2014; 47(3): 122-129]
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA; Cell and Immunobiology, and R & D Center for Cancer Therapeutics, National Cancer Center, Goyang 410-769, Korea
| |
Collapse
|
195
|
Zhang Y, Lima CF, Rodrigues LR. Anticancer effects of lactoferrin: underlying mechanisms and future trends in cancer therapy. Nutr Rev 2014; 72:763-73. [PMID: 25406879 DOI: 10.1111/nure.12155] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Lactoferrin has been widely studied over the last 70 years, and its role in diverse biological functions is now well known and generally accepted by the scientific community. Usually, alterations of the lactoferrin gene in cells are associated with an increased incidence of cancer. Several studies suggest that exogenous treatment with lactoferrin and its derivatives can efficiently inhibit the growth of tumors and reduce susceptibility to cancer. None of these studies, however, reported a consistent outcome with regard to the mechanisms underlying the anticancer effects of lactoferrin. In this review, the association of lactoferrin with cancer is thoroughly discussed, from lactoferrin gene expression to the potential use of lactoferrin in cancer therapy. Lactoferrin cytotoxicity against several cancers is reported to occur in distinct ways under different conditions, namely by cell membrane disruption, apoptosis induction, cell cycle arrest, and cell immunoreaction. Based on these mechanisms, new strategies to improve the anticancer effects of the lactoferrin protein and/or its derivatives are proposed. The potential for lactoferrin in the field of cancer research (including as a chemotherapeutic agent in cancer therapy) is also discussed.
Collapse
Affiliation(s)
- Yunlei Zhang
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal; College of Life Science, Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology, Hunan Normal University, Changsha, China
| | | | | |
Collapse
|
196
|
Svane IM, Verdegaal EM. Achievements and challenges of adoptive T cell therapy with tumor-infiltrating or blood-derived lymphocytes for metastatic melanoma: what is needed to achieve standard of care? Cancer Immunol Immunother 2014; 63:1081-91. [PMID: 25099366 PMCID: PMC11028895 DOI: 10.1007/s00262-014-1580-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/04/2014] [Indexed: 11/28/2022]
Abstract
Adoptive cell therapy (ACT) based on autologous T cell derived either from tumor as tumor-infiltrating lymphocytes (TILs) or from peripheral blood is developing as a key area of future personalized cancer therapy. TIL-based ACT is defined as the infusion of T cells harvested from autologous fresh tumor tissues after ex vivo activation and extensive expansion. TIL-based ACT has so far only been tested in smaller phase I/II studies, but these studies consistently confirm an impressive clinical response rate of up to 50 % in metastatic melanoma including a significant proportion of patients with durable complete tumor eradication. These remarkable results justify the need for a definitive phase III trial documenting the efficacy of this type of T cell-based Advanced Therapy Medicinal Product in order to pave the way for regulatory approval and implementation of TIL therapy as a new treatment standard in oncology practice. TIL-based ACT can, however, only be offered to a limited group of patients based on the need for accessible tumor tissue, the complexity of TIL production procedures, and the very intensive nature of this three-step treatment including both high-dose chemotherapy and interleukin-2 in addition to T cell infusion. To this end, adoptive T cell therapy using peripheral blood mononuclear cell-derived T cells could be a welcome alternative to circumvent these limitations and broaden up the applicability of ACT. Here, we discuss current initiatives in this focused research review.
Collapse
Affiliation(s)
- Inge Marie Svane
- Department of Haematology and Department of Oncology, Herlev Hospital, Center for Cancer Immune Therapy (CCIT), University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark,
| | | |
Collapse
|
197
|
Shi H, Sun M, Liu L, Wang Z. Chimeric antigen receptor for adoptive immunotherapy of cancer: latest research and future prospects. Mol Cancer 2014; 13:219. [PMID: 25241075 PMCID: PMC4177696 DOI: 10.1186/1476-4598-13-219] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/17/2014] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptors (CARs) are recombinant receptors that combine the specificity of an antigen-specific antibody with the T-cell’s activating functions. Initial clinical trials of genetically engineered CAR T cells have significantly raised the profile of T cell therapy, and great efforts have been made to improve this approach. In this review, we provide a structural overview of the development of CAR technology and highlight areas that require further refinement. We also discuss critical issues related to CAR therapy, including the optimization of CAR T cells, the route of administration, CAR toxicity and the blocking of inhibitory molecules.
Collapse
Affiliation(s)
| | | | - Lin Liu
- Department of Oncology, Shandong Cancer Hospital and Institute, No, 440 Jiyan Road, Jinan, Shandong 250117, P,R, China.
| | | |
Collapse
|
198
|
Jensen MC, Riddell SR. Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev 2014; 257:127-44. [PMID: 24329794 DOI: 10.1111/imr.12139] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A major advance in adoptive T-cell therapy (ACT) is the ability to efficiently endow patient's T cells with reactivity for tumor antigens through the stable or regulated introduction of genes that encode high affinity tumor-targeting T-cell receptors (TCRs) or synthetic chimeric antigen receptors (CARs). Case reports and small series of patients treated with TCR- or CAR-modified T cells have shown durable responses in a subset of patients, particularly with B-cell malignancies treated with T cells modified to express a CAR that targets the CD19 molecule. However, many patients do not respond to therapy and serious on and off-target toxicities have been observed with TCR- and CAR-modified T cells. Thus, challenges remain to make ACT with gene-modified T cells a reproducibly effective and safe therapy and to expand the breadth of patients that can be treated to include those with common epithelial malignancies. This review discusses research topics in our laboratories that focus on the design and implementation of ACT with CAR-modified T cells. These include cell intrinsic properties of distinct T-cell subsets that may facilitate preparing therapeutic T-cell products of defined composition for reproducible efficacy and safety, the design of tumor targeting receptors that optimize signaling of T-cell effector functions and facilitate tracking of migration of CAR-modified T cells in vivo, and novel CAR designs that have alternative ligand binding domains or confer regulated function and/or survival of transduced T cells.
Collapse
Affiliation(s)
- Michael C Jensen
- Seattle Children's Research Institute, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
199
|
Abstract
Recent clinical success has underscored the potential for immunotherapy based on the adoptive cell transfer (ACT) of engineered T lymphocytes to mediate dramatic, potent, and durable clinical responses. This success has led to the broader evaluation of engineered T-lymphocyte-based adoptive cell therapy to treat a broad range of malignancies. In this review, we summarize concepts, successes, and challenges for the broader development of this promising field, focusing principally on lessons gleaned from immunological principles and clinical thought. We present ACT in the context of integrating T-cell and tumor biology and the broader systemic immune response.
Collapse
Affiliation(s)
- Marco Ruella
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
200
|
Liu Z, Li Z. Molecular imaging in tracking tumor-specific cytotoxic T lymphocytes (CTLs). Am J Cancer Res 2014; 4:990-1001. [PMID: 25157278 PMCID: PMC4142291 DOI: 10.7150/thno.9268] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/30/2014] [Indexed: 01/15/2023] Open
Abstract
Despite the remarkable progress of adoptive T cell therapy in cancer treatment, there remains an urgent need for the noninvasive tracking of the transfused T cells in patients to determine their biodistribution, viability, and functionality. With emerging molecular imaging technologies and cell-labeling methods, noninvasive in vivo cell tracking is experiencing impressive progress toward revealing the mechanisms and functions of these cells in real time in preclinical and clinical studies. Such cell tracking methods have an important role in developing effective T cell therapeutic strategies and steering decision-making process in clinical trials. On the other hand, they could provide crucial information to accelerate the regulatory approval process on the T cell therapy. In this review, we revisit the advances in tracking the tumor-specific CTLs, highlighting the latest development in human studies and the key challenges.
Collapse
|