1
|
Sarivalasis A, Morotti M, Mulvey A, Imbimbo M, Coukos G. Cell therapies in ovarian cancer. Ther Adv Med Oncol 2021; 13:17588359211008399. [PMID: 33995591 PMCID: PMC8072818 DOI: 10.1177/17588359211008399] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most important cause of gynecological cancer-related mortality. Despite improvements in medical therapies, particularly with the incorporation of drugs targeting homologous recombination deficiency, EOC survival rates remain low. Adoptive cell therapy (ACT) is a personalized form of immunotherapy in which autologous lymphocytes are expanded, manipulated ex vivo, and re-infused into patients to mediate cancer rejection. This highly promising novel approach with curative potential encompasses multiple strategies, including the adoptive transfer of tumor-infiltrating lymphocytes, natural killer cells, or engineered immune components such as chimeric antigen receptor (CAR) constructs and engineered T-cell receptors. Technical advances in genomics and immuno-engineering have made possible neoantigen-based ACT strategies, as well as CAR-T cells with increased cell persistence and intratumoral trafficking, which have the potential to broaden the opportunity for patients with EOC. Furthermore, dendritic cell-based immunotherapies have been tested in patients with EOC with modest but encouraging results, while the combination of DC-based vaccination as a priming modality for other cancer therapies has shown encouraging results. In this manuscript, we provide a clinically oriented historical overview of various forms of cell therapies for the treatment of EOC, with an emphasis on T-cell therapy.
Collapse
Affiliation(s)
- Apostolos Sarivalasis
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matteo Morotti
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Arthur Mulvey
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Martina Imbimbo
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- CHUV, Rue du Bugnon 46, Lausanne BH09-701, Switzerland
| |
Collapse
|
2
|
CD137 + T-Cells: Protagonists of the Immunotherapy Revolution. Cancers (Basel) 2021; 13:cancers13030456. [PMID: 33530328 PMCID: PMC7866028 DOI: 10.3390/cancers13030456] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/23/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary The CD137 receptor is expressed by activated antigen-specific T-cells. CD137+ T-cells were identified inside TILs and PBMCs of different tumor types and have proven to be the naturally occurring antitumor effector cells, capable of expressing a wide variability in terms of TCR specificity against both shared and neoantigenic tumor-derived peptides. The aim of this review is thus summarizing and highlighting their role as drivers of patients’ immune responses in anticancer therapies as well as their potential role in future and current strategies of immunotherapy. Abstract The CD137 receptor (4-1BB, TNF RSF9) is an activation induced molecule expressed by antigen-specific T-cells. The engagement with its ligand, CD137L, is capable of increasing T-cell survival, proliferation, and cytokine production. This allowed to identify the CD137+ T-cells as the real tumor-specific activated T-cell population. In fact, these cells express various TCRs that are specific for a wide range of tumor-derived peptides, both shared and neoantigenic ones. Moreover, their prevalence in sites close to the tumor and their unicity in killing cancer cells both in vitro and in vivo, raised particular interest in studying their potential role in different strategies of immunotherapy. They indeed showed to be a reliable marker able to predict patient’s outcome to immune-based therapies as well as monitor their response. In addition, the possibility of isolating and expanding this population, turned promising in order to generate effector antitumor T-cells in the context of adoptive T-cell therapies. CD137-targeting monoclonal antibodies have already shown their antitumor efficacy in cancer patients and a number of clinical trials are thus ongoing to test their possible introduction in different combination approaches of immunotherapy. Finally, the intracellular domain of the CD137 receptor was introduced in the anti-CD19 CAR-T cells that were approved by FDA for the treatment of pediatric B-cell leukemia and refractory B-cell lymphoma.
Collapse
|
3
|
Kandalaft LE, Odunsi K, Coukos G. Immune Therapy Opportunities in Ovarian Cancer. Am Soc Clin Oncol Educ Book 2021; 40:1-13. [PMID: 32412818 DOI: 10.1200/edbk_280539] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as a highly promising approach in the treatment of epithelial ovarian cancer (EOC). Immune checkpoint blockade (ICB) therapy, PARP inhibitors (PARPis), neoantigen vaccines, and personalized T-cell therapy have been associated with encouraging clinical activity in a small subset of patients. To increase the proportion of patients who are likely to derive benefit, it will be important not only to generate sufficient numbers of antitumor T cells but also to overcome multiple inhibitory networks in the ovarian tumor microenvironment (TME). Therefore, a major direction is to develop biomarkers that would predict responsiveness to different types of immunotherapies and allow treatment selection based on the results. Moreover, such biomarkers would allow rational combination of immunotherapies while minimizing toxicities. In this review, we provide progress on immune therapies and future directions for maximally exploiting immune-based strategies for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY.,Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
4
|
Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019; 37:2460-2471. [PMID: 31403857 DOI: 10.1200/jco.19.00508] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Zhang W, Liu K, Ye B, Hu G, Zhao K, Ren Y, Liang W. Clinical and biological effects of tumor-associated lymphocytes in the presence or absence of chemotherapy for malignant ascites in ovarian cancer patients. Oncol Lett 2017; 14:3379-3386. [PMID: 28927091 PMCID: PMC5587970 DOI: 10.3892/ol.2017.6635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/07/2017] [Indexed: 11/26/2022] Open
Abstract
Tumor-associated lymphocytes (TALs) have been successfully isolated from ascites and solid tumors, however the clinical use of TALs in treating ovarian cancer (OC) has not yet been reported. The present study investigated the efficacy and toxicity of TALs in the presence or absence of chemotherapy in OC patients with malignant ascites (MA). A total of 32 patients were enrolled in this study. A total of 8 patients received treatment with TALs alone (TALs group), 11 patients received combined treatment of TALs and chemotherapy (combination group) and 13 patients received chemotherapy alone (chemotherapy group). The endpoints included Karnofsky performance status (KPS), ascites-associated symptoms (AAS), time to progression (TTP) and overall survival (OS). Compared with the TALs and chemotherapy group, the KPS and AAS in the combination group significantly improved following treatment. Patients in the TALs group (37.5%) and chemotherapy group (53.8%) achieved significantly fewer objective response rates of ascites compared with those in the combination group (90.9%). Furthermore, combination therapy significantly extended TTP (13 months) compared with TALs alone (1 month, P<0.001), and chemotherapy alone (6 months, P=0.027). Similar results were observed for OS between the combination group and the TALs group (25 vs. 7 months, P<0.001). The present study therefore demonstrates that combined therapy of TALs and chemotherapy is safe, feasible, and more effective than chemotherapy or TALs alone in controlling MA and improving the quality of life for OC patients.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Medical Oncology, The Sixth People's Hospital of Chengdu, Chengdu, Sichuan 610051, P.R. China.,Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Kejun Liu
- Department of Medical Oncology, Dongguan People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Bin Ye
- Department of Medical Oncology, The Sixth People's Hospital of Chengdu, Chengdu, Sichuan 610051, P.R. China
| | - Guolin Hu
- Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Kelei Zhao
- Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yazhou Ren
- Big Data Research Center, School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, P.R. China
| | - Weijiang Liang
- Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
6
|
Nayama M, Collinet P, Salzet M, Vinatier D. [Immunological aspects of ovarian cancer: Therapeutic perspectives]. ACTA ACUST UNITED AC 2016; 45:1020-1036. [PMID: 27320132 DOI: 10.1016/j.jgyn.2016.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 05/07/2016] [Accepted: 05/13/2016] [Indexed: 01/09/2023]
Abstract
Ovarian cancer is recognized by the immunological system of its host. Initially, it is effective to destroy and eliminate the cancer. But gradually, resistant tumor cells more aggressive and those able to protect themselves by inducing immune tolerance will be selected. Immunotherapy to be effective should consider both components of immune response with an action on cytotoxic immune effectors and action on tolerance mechanisms. The manipulations of the immune system should be cautious, because the immune effects are not isolated. A theoretically efficient handling may simultaneously cause an adverse effect which was not envisaged and could neutralize the benefits of treatment. Knowledge of tolerance mechanisms set up by the tumor is for the clinician a prerequisite before they prescribe these treatments. For each cancer, the knowledge of its immunological status is a prerequisite to propose adapted immunological therapies.
Collapse
Affiliation(s)
- M Nayama
- Service de gynécologie obstétrique, maternité Issaka-Gazoby, BP 10975, Niamey, Niger
| | - P Collinet
- CHU de Lille, 59000 Lille, France; Département universitaire de gynécologie obstétrique, université Nord-de-France, 59045 Lille cedex, France
| | - M Salzet
- EA 4550, IFR 147, laboratoire PRISM : protéomique, réponse inflammatoire, spectrométrie de Masse, université Lille 1, bâtiment SN3, 1(er) étage, 59655 Villeneuve d'Ascq cedex, France
| | - D Vinatier
- CHU de Lille, 59000 Lille, France; EA 4550, IFR 147, laboratoire PRISM : protéomique, réponse inflammatoire, spectrométrie de Masse, université Lille 1, bâtiment SN3, 1(er) étage, 59655 Villeneuve d'Ascq cedex, France; Département universitaire de gynécologie obstétrique, université Nord-de-France, 59045 Lille cedex, France.
| |
Collapse
|
7
|
Geukes Foppen MH, Donia M, Svane IM, Haanen JBAG. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol Oncol 2015; 9:1918-35. [PMID: 26578452 DOI: 10.1016/j.molonc.2015.10.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/15/2022] Open
Abstract
Over the past few years melanoma incidence has been rising steadily, resulting in an increase in melanoma related mortality. Until recently, therapeutic options for metastatic melanoma were scarce. Chemotherapy and, in some countries, IL-2 were the only registered treatment modalities. In the last five years, treatment with immunotherapy (anti CTLA-4, anti PD-1, or the combination of these antibodies) has shown very promising results and was able to improve survival in patients with metastatic melanoma. Adoptive cell therapy using tumor-infiltrating lymphocytes is yet another, but highly promising, immunotherapeutic strategy for patients with metastatic melanoma. This review will discuss the development of TIL as a treatment option for melanoma, its mode of action and simplification over time, and the possibilities to expand this therapy to other types of cancer. Also, the future directions of TIL based therapies will be highlighted.
Collapse
Affiliation(s)
- M H Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| | - M Donia
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - I M Svane
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - J B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
8
|
B7-H4 as a potential target for immunotherapy for gynecologic cancers: a closer look. Gynecol Oncol 2014; 134:181-189. [PMID: 24657487 DOI: 10.1016/j.ygyno.2014.03.553] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/14/2014] [Accepted: 03/16/2014] [Indexed: 12/16/2022]
Abstract
B7-H4 is a transmembrane protein that binds an unknown receptor on activated T cells resulting in inhibition of T-cell effector function via cell cycle arrest, decreased proliferation, and reduced IL-2 production. B7-H4 is up-regulated on the surface of cancer cells and immunosuppressive tumor-associated macrophages (TAMs) in a variety of human cancers. Notably, B7-H4 expression levels inversely correlate with patient survival in ovarian cancer, making B7-H4 an attractive candidate for therapeutic intervention. Here, we summarize the experimental data and methodologies that have revealed B7-H4's mRNA and protein expression and function in both mice and humans since its discovery in 2003, with a specific focus on B7-H4's role in ovarian cancer. We also underscore the discrepancies in published data due to high variability in methodology and use of different antibodies, most of which are not commercially available. Finally, since B7-H4 is expressed on tumor cells and TAMs in various cancer types, directing therapeutics against B7-H4 could have tremendous synergistic outcomes in favorably altering the tumor micro-environment and eliminating cancer cells. We highlight the therapeutic potential of targeting B7-H4, both by comparing other negative immune modulators such as PD-1 and CTLA-4 and by identifying novel methods to target B7-H4 directly or indirectly to overcome B7-H4-mediated T-cell inhibition.
Collapse
|
9
|
Ye Q, Song DG, Poussin M, Yamamoto T, Best A, Li C, Coukos G, Powell DJ. CD137 accurately identifies and enriches for naturally occurring tumor-reactive T cells in tumor. Clin Cancer Res 2013; 20:44-55. [PMID: 24045181 DOI: 10.1158/1078-0432.ccr-13-0945] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE Upregulation of CD137 (4-1BB) on recently activated CD8(+) T cells has been used to identify rare viral or tumor antigen-specific T cells from peripheral blood. Here, we evaluated the immunobiology of CD137 in human cancer and the utility of a CD137-positive separation methodology for the identification and enrichment of fresh tumor-reactive tumor-infiltrating lymphocytes (TIL) or tumor-associated lymphocytes (TAL) from ascites for use in adoptive immunotherapy. EXPERIMENTAL DESIGN TILs from resected ovarian cancer or melanoma were measured for surface CD137 expression directly or after overnight incubation in the presence of tumor cells and homeostatic cytokines. CD137(pos) TILs were sorted and evaluated for antitumor activity in vitro and in vivo. RESULTS Fresh ovarian TILs and TALs naturally expressed higher levels of CD137 than circulating T cells. An HLA-dependent increase in CD137 expression was observed following incubation of fresh enzyme-digested tumor or ascites in IL-7 and IL-15 cytokines, but not IL-2. Enriched CD137(pos) TILs, but not PD-1(pos) or PD-1(neg) CD137(neg) cells, possessed autologous tumor reactivity in vitro and in vivo. In melanoma studies, all MART-1-specific CD8(+) TILs upregulated CD137 expression after incubation with HLA-matched, MART-expressing cancer cells and antigen-specific effector function was restricted to the CD137(pos) subset in vitro. CD137(pos) TILs also mediated superior antitumor effects in vivo, compared with CD137(neg) TILs. CONCLUSIONS Our findings reveal a role for the TNFR-family member CD137 in the immunobiology of human cancer where it is preferentially expressed on tumor-reactive subset of TILs, thus rationalizing its agonistic engagement in vivo and its use in TIL selection for adoptive immunotherapy trials.
Collapse
Affiliation(s)
- Qunrui Ye
- Authors' Affiliations: Department of Obstetrics and Gynecology, Ovarian Cancer Research Center; and Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Bernal SD, Ona ET, Riego-Javier A, DE Villa R, Cristal-Luna GR, Laguatan JB, Batac ER, Canlas OQ. Anticancer immune reactivity and long-term survival after treatment of metastatic ovarian cancer with dendritic cells. Oncol Lett 2011; 3:66-74. [PMID: 22740858 DOI: 10.3892/ol.2011.424] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 08/26/2011] [Indexed: 01/02/2023] Open
Abstract
Hematopoietic stem cells collected by leukapheresis of a patient with metastatic ovarian carcinoma (OVCA) were induced into dendritic cell (DC) differentiation and fused with liposomal constructs of autologous and allogeneic ovarian carcinoma antigens (DC-OVCA). The proliferation of autologous T cells induced by DCs was determined by [(3)H]-thymidine uptake. Maximal T-cell proliferation was observed in co-cultures of DCs fused with liposomal OVCA constructs compared with intact autologous OVCA cells. The combination of autologous and allogeneic liposomal OVCA constructs induced greater T-cell proliferation than either alone. The cytotoxicity of DC-activated T cells against various target cells were analyzed by a (51)Cr-release assay. The combination of autologous and allogeneic liposomal OVCA constructs showed the highest stimulation of T cell-mediated cytotoxicity against OVCA cells, but had minimal cytotoxicity against normal fibroblasts or leukemia cells. The liposomal preparations of DC-OVCA were injected monthly into a patient with metastatic ovarian carcinoma whose tumors progressed following multiple courses of chemotherapy. DCs analyzed from the patient post-immunization showed 2- to 3-fold greater OVCA cytotoxicity compared to pre-immunization DCs. Immunoblots using the patient's serum showed reactivity with a number of proteins from ovarian cancer extracts, but not in normal fibroblasts and breast cancer. Following the DC-OVCA treatment, the metastatic lesions progressively decreased in size to the point of being undetectable by serial CAT scans. Seven years following the initial diagnosis, the patient continues to be free of cancer. This report described the anticancer immune reactivity and anti-tumor response induced by DCs sensitized with liposomal constructs of OVCA antigens. Immune cell therapy may therefore be a useful adjunct to surgery and chemotherapy for the treatment of ovarian cancer.
Collapse
|
11
|
Tumor immune surveillance and ovarian cancer: lessons on immune mediated tumor rejection or tolerance. Cancer Metastasis Rev 2011; 30:141-51. [PMID: 21298574 DOI: 10.1007/s10555-011-9289-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the past few years, cancer immunotherapies have produced promising results. Although traditionally considered unresponsive to immune therapy, increasing evidence indicates that ovarian cancers are, in fact, immunogenic tumors. This evidence comes from diverse epidemiologic and clinical data comprising evidence of spontaneous antitumor immune response and its association with longer survival in a proportion of ovarian cancer patients; evidence of tumor immune evasion mechanisms and their association with short survival in some ovarian cancer patients; and finally pilot data supporting the efficacy of immune therapy. Below we will discuss lessons learned on the biology underlying ovarian cancer immune rejection or tolerance and we will discuss its association with clinical outcome. We will discuss the role of angiogenesis and the tumor endothelium on regulation of the antitumor immune response with a special emphasis on the role of vascular endothelial growth factor (VEGF) in the suppression of immunological processes, which control tumor progression and its unique crosstalk with endothelin systems, and how their interactions may shape the antitumor immune response. In addition, we will discuss mechanisms of tumor tolerance through the suppression or exhaustion of effector cells and how these could be countered in the clinic. We believe that understanding these pathways in the tumor microenvironment will lead to novel strategies for enhancing ovarian cancer immunotherapy.
Collapse
|
12
|
Cheng M, Ma J, Chen Y, Zhang J, Zhao W, Zhang J, Wei H, Ling B, Sun R, Tian Z. Establishment, characterization, and successful adaptive therapy against human tumors of NKG cell, a new human NK cell line. Cell Transplant 2011; 20:1731-46. [PMID: 21669033 DOI: 10.3727/096368911x580536] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Natural killer (NK) cells play important roles in adoptive cellular immunotherapy against certain human cancers. This study aims to establish a new human NK cell line and to study its role for adoptive cancer immunotherapy. Peripheral blood samples were collected from 54 patients to establish the NK cell line. A new human NK cell line, termed as NKG, was established from a Chinese male patient with rapidly progressive non-Hodgkin's lymphoma. NKG cells showed LGL morphology and were phenotypically identified as CD56(bright) NK cell with CD16(-), CD27(-), CD3(-), αβTCR(-), γδTCR(-), CD4(-), CD8(-), CD19(-), CD161(-), CD45(+), CXCR4(+), CCR7(+), CXCR1(-), and CX3CR1(-). NKG cells showed high expression of adhesive molecules (CD2, CD58, CD11a, CD54, CD11b, CD11c), an array of activating receptors (NKp30, NKp44, NKp46, NKG2D, NKG2C), and cytolysis-related receptors and molecules (TRAIL, FasL, granzyme B, perforin, IFN-γ). The cytotoxicity of NKG cells against tumor cells was higher than that of the established NK cell lines NK-92, NKL, and YT. NKG cell cytotoxicity depended on the presence of NKG2D and NKp30. When irradiated with 8 Gy, NKG cells were still with high cytotoxicity and activity in vitro and with safety in vivo, but without proliferation. Further, the irradiated NKG cells exhibited strong cytotoxicity against human primary ovarian cancer cells in vitro, and against human ovarian cancer in a mouse xenograft model. The adoptive transfer of NKG cells significantly inhibited the ovarian tumor growth, decreased the mortality rate and prolonged the survival, even in cases of advanced diseases. A number of NKG cells were detected in the ovarian tumor tissues during cell therapy. In use of the new human NK cell line, NKG would a promising cellular candidate for adoptive immunotherapy of human cancer.
Collapse
Affiliation(s)
- Min Cheng
- Institute of Immunology, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Thibodeaux SR, Curiel TJ. Immune Therapy for Ovarian Cancer: Promise and Pitfalls. Int Rev Immunol 2011; 30:102-19. [DOI: 10.3109/08830185.2011.567361] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
14
|
Adams SF, Levine DA, Cadungog MG, Hammond R, Facciabene A, Olvera N, Rubin SC, Boyd J, Gimotty PA, Coukos G. Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer 2009; 115:2891-902. [PMID: 19472394 DOI: 10.1002/cncr.24317] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The aim of the study was to determine whether tumor-infiltrating lymphocytes and/or tumor mitotic activity could identify subgroups of patients with advanced serous epithelial ovarian cancer who would maximally benefit from aggressive surgical cytoreduction. METHODS Snap-frozen specimens from 134 consecutive patients with stage III or IV serous or poorly differentiated ovarian adenocarcinoma undergoing primary debulking surgery from a single US institution were characterized based on CD3(+), CD8(+), FoxP3(+) tumor-infiltrating lymphocytes, and Ki67 expression. Kaplan-Meier survival curves were estimated and compared using a log-rank statistic. A multivariate Cox model was used to estimate adjusted hazard ratios. Interactions were modeled using recursive partitioning based on maximal prognostic differentiation. RESULTS Brisk intraepithelial CD8(+) cells (P = .035) and low Ki67 expression (P = .042) portended prolonged survival. The T-cell infiltration was more likely to occur in tumors with high proliferation index. Patients whose tumors exhibited low Ki67 expression and high intraepithelial CD8(+) frequency had a 5-year survival rate of 73.3%. Patients with aggressive tumor behavior, that is, whose tumors exhibited low frequency of intraepithelial CD8(+) T cells or high Ki67 expression were more likely to draw benefit from aggressive surgical cytoreduction. Survival was similar for patients with brisk CD8(+) T cells who had optimal or suboptimal debulking. Likewise, survival was similar for patients with low Ki67 expression who had optimal or suboptimal debulking. CONCLUSIONS For the first time, these novel interactions of T cells, tumor proliferation index, and surgical treatment reveal that biological prognosticators may be useful for surgical decision making in ovarian cancer.
Collapse
Affiliation(s)
- Sarah F Adams
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
BACKGROUND Ovarian cancer is frequently diagnosed at an advanced stage, and although initially responsive to surgery and chemotherapy, has a high rate of recurrence and mortality. Cellular immunotherapy may offer the prospect of treatment to prevent or delay recurrent metastatic disease. OBJECTIVE To provide an overview of current innovations in cellular immunotherapy for ovarian cancer, with an emphasis on dendritic cell vaccination and adoptive T-cell immunotherapy. METHODS Three key areas are explored in this review: first, an appraisal of the current state of the art of cellular immunotherapy for treatment of ovarian cancer; second, a discussion of the immunological defenses erected by ovarian cancer to prevent immunological attack, with an emphasis on the role of tumor-associated regulatory T cells; and third, an exploration of innovative techniques that may enhance the ability of cellular immunotherapy to overcome ovarian tumor-associated immune suppression. RESULTS/CONCLUSION Ovarian cancer is recognized as a paradigm for tumor-associated immune suppression. Innovative approaches for antagonism of tumor-associated regulatory T-cell infiltration and redirection of self antigen-driven regulatory T-cell activation may provide the key to development of future strategies for cellular immunotherapy against ovarian cancer.
Collapse
Affiliation(s)
- Martin J Cannon
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, 4301 West Markham, Little Rock, AR 72205, USA.
| | | |
Collapse
|
16
|
Abstract
Ovarian cancer remains a challenging disease for which improved treatments are urgently needed. Most patients present with advanced disease that is highly responsive to surgery combined with platinum- and taxane-based chemotherapy, with a state of minimal residual disease being achieved in many cases. However, chemotherapy-resistant recurrent tumors typically appear within 1-5 years and are ultimately fatal. Recently, several groups have shown that ovarian tumors are often infiltrated by activated T cells at the time of diagnosis, and patients with dense infiltrates of CD3+CD8+ T cells experience unexpectedly favorable progression-free and overall survival. Other cell types in the immune infiltrate oppose anti-tumor immunity, including CD4+CD25+FoxP3+ regulatory T cells, CD8+ regulatory T cells, macrophages, and dendritic cells. The composition of immune infiltrates is shaped by the expression of cytokines, chemokines, antigens, major histocompatibility complex molecules, and costimulatory molecules. The relationship between these various immunological factors is reviewed here with a strong emphasis on outcomes data so as to create a knowledge base that is well grounded in clinical reality. With improved understanding of the functional properties of natural CD8+ T-cell responses to ovarian cancer, there is great potential to improve clinical outcomes by amplifying host immunity.
Collapse
Affiliation(s)
- Brad H Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC, Canada.
| |
Collapse
|
17
|
Tumor-infiltrating T cells correlate with NY-ESO-1-specific autoantibodies in ovarian cancer. PLoS One 2008; 3:e3409. [PMID: 18923710 PMCID: PMC2561074 DOI: 10.1371/journal.pone.0003409] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 09/17/2008] [Indexed: 12/20/2022] Open
Abstract
Background Tumor-infiltrating CD8+ T cells are correlated with prolonged progression-free and overall survival in epithelial ovarian cancer (EOC). A significant fraction of EOC patients mount autoantibody responses to various tumor antigens, however the relationship between autoantibodies and tumor-infiltrating T cells has not been investigated in EOC or any other human cancer. We hypothesized that autoantibody and T cell responses may be correlated in EOC and directed toward the same antigens. Methodology and Principal Findings We obtained matched serum and tumor tissue from 35 patients with high-grade serous ovarian cancer. Serum samples were assessed by ELISA for autoantibodies to the common tumor antigen NY-ESO-1. Tumor tissue was examined by immunohistochemistry for expression of NY-ESO-1, various T cell markers (CD3, CD4, CD8, CD25, FoxP3, TIA-1 and Granzyme B) and other immunological markers (CD20, MHC class I and MHC class II). Lymphocytic infiltrates varied widely among tumors and included cells positive for CD3, CD8, TIA-1, CD25, FoxP3 and CD4. Twenty-six percent (9/35) of patients demonstrated serum IgG autoantibodies to NY-ESO-1, which were positively correlated with expression of NY-ESO-1 antigen by tumor cells (r = 0.57, p = 0.0004). Autoantibodies to NY-ESO-1 were associated with increased tumor-infiltrating CD8+, CD4+ and FoxP3+ cells. In an individual HLA-A2+ patient with autoantibodies to NY-ESO-1, CD8+ T cells isolated from solid tumor and ascites were reactive to NY-ESO-1 by IFN-γ ELISPOT and MHC class I pentamer staining. Conclusion and Significance We demonstrate that tumor-specific autoantibodies and tumor-infiltrating T cells are correlated in human cancer and can be directed against the same target antigen. This implies that autoantibodies may collaborate with tumor-infiltrating T cells to influence clinical outcomes in EOC. Furthermore, serological screening methods may prove useful for identifying clinically relevant T cell antigens for immunotherapy.
Collapse
|
18
|
Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer. Expert Rev Anticancer Ther 2008; 8:243-57. [PMID: 18279065 DOI: 10.1586/14737140.8.2.243] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is responsible for the majority of gynecologic cancer deaths and despite the highest standard of multimodality therapy with surgery and cytotoxic chemotherapy, long-term survival remains low. With compelling evidence that epithelial ovarian cancer is an immunogenic tumor capable of stimulating an antitumor immune response, renewed efforts to develop immune therapies to augment the efficacy of traditional therapies are underway. Current immunotherapies focus on varied modes of antitumor vaccine development, particularly with the use of dendritic cell vaccines, effective methods for adoptive T-cell transfer and combinatorial approaches with immune modulatory therapy subverting natural tolerance mechanisms or boosting effector mechanisms. Additional combinatorial approaches include the use of cytokines and/or chemotherapy with immune therapy.
Collapse
Affiliation(s)
- Christina S Chu
- University of Pennsylvania, Division of Gynecologic Oncology, Center for Research on Ovarian Cancer, PA, USA.
| | | | | | | |
Collapse
|
19
|
Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006; 12:6106-15. [PMID: 17062687 PMCID: PMC2154351 DOI: 10.1158/1078-0432.ccr-06-1183] [Citation(s) in RCA: 937] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE A phase I study was conducted to assess the safety of adoptive immunotherapy using gene-modified autologous T cells for the treatment of metastatic ovarian cancer. EXPERIMENTAL DESIGN T cells with reactivity against the ovarian cancer-associated antigen alpha-folate receptor (FR) were generated by genetic modification of autologous T cells with a chimeric gene incorporating an anti-FR single-chain antibody linked to the signaling domain of the Fc receptor gamma chain. Patients were assigned to one of two cohorts in the study. Eight patients in cohort 1 received a dose escalation of T cells in combination with high-dose interleukin-2, and six patients in cohort 2 received dual-specific T cells (reactive with both FR and allogeneic cells) followed by immunization with allogeneic peripheral blood mononuclear cells. RESULTS Five patients in cohort 1 experienced some grade 3 to 4 treatment-related toxicity that was probably due to interleukin-2 administration, which could be managed using standard measures. Patients in cohort 2 experienced relatively mild side effects with grade 1 to 2 symptoms. No reduction in tumor burden was seen in any patient. Tracking 111In-labeled adoptively transferred T cells in cohort 1 revealed a lack of specific localization of T cells to tumor except in one patient where some signal was detected in a peritoneal deposit. PCR analysis showed that gene-modified T cells were present in the circulation in large numbers for the first 2 days after transfer, but these quickly declined to be barely detectable 1 month later in most patients. An inhibitory factor developed in the serum of three of six patients tested over the period of treatment, which significantly reduced the ability of gene-modified T cells to respond against FR+ tumor cells. CONCLUSIONS Large numbers of gene-modified tumor-reactive T cells can be safely given to patients, but these cells do not persist in large numbers long term. Future studies need to employ strategies to extend T cell persistence. This report is the first to document the use of genetically redirected T cells for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Michael H. Kershaw
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Cancer Immunology Research Program, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Australia
- Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Jennifer A. Westwood
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Cancer Immunology Research Program, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Australia
| | - Linda L. Parker
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Gang Wang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Zelig Eshhar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon A. Mavroukakis
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Donald E. White
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - John R. Wunderlich
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Linda Rogers-Freezer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Clara C. Chen
- Department of Nuclear Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - James C. Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Steven A. Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Patrick Hwu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Coukos G, Conejo-Garcia JR, Roden RBS, Wu TC. Immunotherapy for gynaecological malignancies. Expert Opin Biol Ther 2005; 5:1193-210. [PMID: 16120050 DOI: 10.1517/14712598.5.9.1193] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Gynaecological malignancies, excluding breast cancer, cause approximately 25,000 deaths yearly among women in the US. Therefore, novel approaches for the prevention or treatment of these diseases are urgently required. In the case of cervical cancer, human papillomavirus (HPV) xenoantigens are readily recognised by the immune system, and their targeting has shown great promise in preclinical models of therapeutic vaccination and in clinical studies of preventative vaccination. A growing body of evidence indicates that ovarian cancer is also immunogenic and can thus be targeted through immunotherapy. This review outlines the principles and problems of immunotherapy for cervical and ovarian cancer, including the authors' personal assessment.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Heterophile/immunology
- Antigens, Neoplasm/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Cytokines
- Drug Evaluation, Preclinical
- Female
- Genital Neoplasms, Female/immunology
- Genital Neoplasms, Female/prevention & control
- Genital Neoplasms, Female/therapy
- Humans
- Immunotherapy/methods
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/prevention & control
- Ovarian Neoplasms/therapy
- Papillomaviridae/immunology
- T-Lymphocytes, Regulatory
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/prevention & control
- Uterine Cervical Neoplasms/therapy
- Vaccines, Attenuated/therapeutic use
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- George Coukos
- Abramson Cancer Research Institute, Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
21
|
Nijman HW, Lambeck A, van der Burg SH, van der Zee AGJ, Daemen T. Immunologic aspect of ovarian cancer and p53 as tumor antigen. J Transl Med 2005; 3:34. [PMID: 16164749 PMCID: PMC1243238 DOI: 10.1186/1479-5876-3-34] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Accepted: 09/15/2005] [Indexed: 01/13/2023] Open
Abstract
Ovarian cancer represents the fifth leading cause of death from all cancers for women. During the last decades overall survival has improved due to the use of new chemotherapy schedules. Still, the majority of patients die of this disease. Research reveals that ovarian cancer patients exhibit significant immune responses against their tumor. In this review the knowledge obtained thus far on the interaction of ovarian cancer tumor cells and the immune system is discussed. Furthermore the role of p53 as tumor antigen and its potential role as target antigen in ovarian cancer is summarized. Based on the increased knowledge on the role of the immune system in ovarian cancer major improvements are to be expected of immunotherapy based treatment of this disease.
Collapse
Affiliation(s)
- HW Nijman
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
| | - A Lambeck
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
- Dept. of Medical Microbiology, Molecular Virology Section, Groningen University Medical Center
| | - SH van der Burg
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - AGJ van der Zee
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
| | - T Daemen
- Dept. of Medical Microbiology, Molecular Virology Section, Groningen University Medical Center
| |
Collapse
|
22
|
Yu R, Fujio K, Tahara H, Araki Y, Yamamoto K. Clonal dynamics of tumor-infiltrating lymphocytes. Eur J Immunol 2005; 35:1754-63. [PMID: 15902685 DOI: 10.1002/eji.200425866] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The presence of tumor-infiltrating lymphocytes (TIL) provides important evidence of anti-tumor immunity in vivo. However, TIL are usually not sufficient for inhibiting tumor growth. We explored the spatial and temporal aspects of clonal accumulation of TIL using RT-PCR/single-strand conformation polymorphism analysis. In CMS5 fibrosarcomas in BALB/c mice, accumulated T cell clones were specific in that dominant TIL were identical between distant tumors. Moreover, dominant TIL in the first tumor appeared consistently in the second tumor inoculated after formation of the first tumor. These results suggest that TIL show a certain level of specific tumor surveillance. When we characterized CD4(+) and CD8(+) TIL separately, CD8(+) TIL were highly concentrated and persistently localized at the tumor site, while most CD4(+) TIL clones were less concentrated and less persistent. A functional analysis showed that TIL had a certain degree of anti-tumor activity when CD4(+) and CD8(+) TIL were co-transferred. Co-transfer of CD4(+) and CD8(+) TIL exhibited equivalent anti-tumor activity, irrespective of tumor stage. However, the numbers of TIL did not increase after the early phase of tumor progression. These data suggest that TIL are specific to the tumor and potentially retain anti-tumor activity, although their accumulation in mice is impaired.
Collapse
Affiliation(s)
- Rong Yu
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
23
|
Monos DS, Pappas J, Magira EE, Gaughan J, Aplenc R, Sakkas L, Freedman R, Reveille JD, Platsoucas CD. Identification of HLA-DQα and -DRβ Residues Associated With Susceptibility and Protection to Epithelial Ovarian Cancer. Hum Immunol 2005; 66:554-62. [PMID: 15935893 DOI: 10.1016/j.humimm.2005.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 01/19/2005] [Indexed: 11/30/2022]
Abstract
Substantial evidence has been accumulated suggesting that T cells in patients with epithelial ovarian carcinoma (EOC) exhibit an antigen-driven immune response directed against the tumor cells. In the context of human leukocyte antigen (HLA), this suggests its possible involvement in the disease. Therefore, we examined the distribution of the HLA-DRB1*, -DQA1*, and -DQB1* alleles in 47 patients with EOC and 67 healthy Caucasian women. The frequency of D(70) and E(71) polymorphic residues of the DRB1 alleles was significantly reduced in EOC patients versus controls (pD(70)E(71) = 0.009), suggesting a protective role against the disease. The DQalpha residues R(52) and Y(11)R(55) were increased in the patients (p = 0.008 and 0.012, respectively). Because residues 11 and 55 participate in the formation of pocket 1, they may be functionally important amino acid positions that influence disease susceptibility. The frequency of the DQalpha susceptibility epitope (R(52)Y(11)R(55)) among the DRbetaD(70)E(71)-positive EOC patients was increased when compared with DRbetaD(70)E(71)-positive controls (EOC, 100%; control, 52%; p = 0.028). Among individuals without the DQalpha susceptibility epitope, the distribution of DRbetaD(70)E(71)-positive cases was significantly different between EOC patients and controls (EOC, 0%; control, 60%; p = 0.039). Therefore, it appears that the presence of DQalpha susceptibility elements overrides the protective effect of the DRbetaD(70)E(71) epitope and suggests an interactive relationship between DRbeta and DQalpha epitopes that may be of importance for disease susceptibility. Because positions DRbeta 70,71 and DQalpha 52 have been implicated in immunologic diseases, it is likely that besides being critical for T-cell recognition, they may also play a role in T-cell development and acquisition of the T-cell repertoire.
Collapse
Affiliation(s)
- Dimitri S Monos
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pappas J, Jung WJ, Barda AK, Lin WL, Fincke JE, Purev E, Radu M, Gaughan J, Helm CW, Hernandez E, Freedman RS, Platsoucas CD. Substantial proportions of identical β-chain T-cell receptor transcripts are present in epithelial ovarian carcinoma tumors. Cell Immunol 2005; 234:81-101. [PMID: 16038891 DOI: 10.1016/j.cellimm.2005.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 05/10/2005] [Accepted: 05/15/2005] [Indexed: 11/28/2022]
Abstract
To determine whether clonally expanded T cells are present in tumor specimens from patients with epithelial ovarian carcinoma (EOC) we amplified by the non-palindromic adaptor PCR (NPA-PCR) or by Vbeta-specific PCR beta-chain T-cell receptor (TCR) transcripts from these tumor specimens. The amplified transcripts were cloned and sequenced. Sequence analysis revealed the presence of substantial proportions of multiple identical copies of beta-chain TCR transcripts, suggesting the presence of clonal expansions of T cells in these patients, which were statistically significant by the binomial distribution in seven of nine patients. Independent amplification in separate experiments of beta-chain TCR transcripts from one patient by either NPA-PCR or by Vbeta-specific PCR, followed by cloning and sequencing, revealed identical clonal expansions irrespectively of the amplification method used. Multiple identical copies of beta-chain TCR transcripts can be derived only by specific antigen-driven proliferation and clonal expansion of the T-cell clones which recognize these antigens. Because of the very large size of the TCR repertoire, the probability of finding by chance multiple identical copies of these transcripts within an independent sample of T cells is negligible. These results demonstrate that T cells infiltrating solid tumor specimens or malignant ascites of patients with EOC contain monoclonal/oligoclonal populations of T cells.
Collapse
Affiliation(s)
- John Pappas
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Helal TEA, Alla AEK, Laban MA, Fahmy RM. Immunophenotyping of tumor-infiltrating mononuclear cells in ovarian carcinoma. Pathol Oncol Res 2004; 10:80-4. [PMID: 15188023 DOI: 10.1007/bf02893460] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Accepted: 04/22/2004] [Indexed: 10/21/2022]
Abstract
Infiltrating mononuclear cells play an important role in many types of cancer. The aim of this work was to determine the immunologic characteristics of mononuclear cellular infiltrate in ovarian cancer as compared to benign ovarian tumors. Paraffin-embedded tissues obtained from 52 ovarian carcinomas and 21 benign ovarian neoplasms were examined immunohistochemically to demonstrate suppressor/cytotoxic T cells and macrophages by using CD8 and CD68 monoclonal antibodies, respectively. The mean percentage of CD8+ cells was much higher in the malignant than in the benign group (P=0.00009). Similarly, the mean level of CD68+ cells was significantly higher in carcinomas than in benign cases (P=0.006). There was a significant negative correlation between the percentage of CD8+ cells and CD68+ cells in the malignant group (P=0.000002). Conversely, no correlation could be obtained between the values of these two cell types in the benign lesions. In the malignant group, although the percentages of CD8+ cells and CD68+ cells were not related to tumor differentiation, they were significantly related to tumor type. CD8+ cells were significantly higher in the serous (P=0.02), and CD68+ cells were higher in the mucinous carcinomas (P=0.0005). CD8+ T cells and macrophages constitute a major component of the infiltrating mononuclear cells in ovarian carcinoma. Their frequency seems to be related to the tumor type rather than the degree of tumor differentiation.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Differentiation
- Cystadenocarcinoma, Mucinous/pathology
- Cystadenocarcinoma, Serous/pathology
- Female
- Humans
- Immunoenzyme Techniques
- Immunophenotyping
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Lymphocytes, Tumor-Infiltrating/pathology
- Macrophages
- Ovarian Neoplasms/pathology
Collapse
Affiliation(s)
- Thanaa el-A Helal
- Department of Pathology, Ain Shams Faculty of Medicine, Cairo, Egypt.
| | | | | | | |
Collapse
|
26
|
Knutson KL, Curiel TJ, Salazar L, Disis ML. Immunologic principles and immunotherapeutic approaches in ovarian cancer. Hematol Oncol Clin North Am 2003; 17:1051-73. [PMID: 12959191 DOI: 10.1016/s0889-8588(03)00064-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ovarian cancer is an immunogenic tumor, and numerous antigens have been identified in recent years. Several of these antigens are important in regulating tumor growth and may be ideal targets for the development of immune-based strategies. In the absence of immunologic intervention, tumors evade the immune system by several mechanisms, most notably tolerance and immunosuppression. As understanding of the immune response improves, strategies are being designed to circumvent T-cell tolerance to self-antigens through modulation of APC function. In addition, techniques are being developed to identify reverse ovarian cancer-induced immune evasion tactics. The type of the immune-based therapy to apply varies with disease burden. It is hoped that discoveries at the bench along with lessons learned in prior clinical trials soon will allow clinicians to develop rationally based immunologic strategies to treat and prevent ovarian cancer.
Collapse
Affiliation(s)
- Keith L Knutson
- Tumor Vaccine Group, 1959 Northeast Pacific Street, Box 356527, HSB BB1321, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
27
|
Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348:203-13. [PMID: 12529460 DOI: 10.1056/nejmoa020177] [Citation(s) in RCA: 2579] [Impact Index Per Article: 117.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Although tumor-infiltrating T cells have been documented in ovarian carcinoma, a clear association with clinical outcome has not been established. METHODS We performed immunohistochemical analysis of 186 frozen specimens from advanced-stage ovarian carcinomas to assess the distribution of tumor-infiltrating T cells and conducted outcome analyses. Molecular analyses were performed in some tumors by real-time polymerase chain reaction. RESULTS CD3+ tumor-infiltrating T cells were detected within tumor-cell islets (intratumoral T cells) in 102 of the 186 tumors (54.8 percent); they were undetectable in 72 tumors (38.7 percent); the remaining 12 tumors (6.5 percent) could not be evaluated. There were significant differences in the distributions of progression-free survival and overall survival according to the presence or absence of intratumoral T cells (P<0.001 for both comparisons). The five-year overall survival rate was 38.0 percent among patients whose tumors contained T cells and 4.5 percent among patients whose tumors contained no T cells in islets. Significant differences in the distributions of progression-free survival and overall survival according to the presence or absence of intratumoral T cells (P<0.001 for both comparisons) were also seen among 74 patients with a complete clinical response after debulking and platinum-based chemotherapy: the five-year overall survival rate was 73.9 percent among patients whose tumors contained T cells and 11.9 percent among patients whose tumors contained no T cells in islets. The presence of intratumoral T cells independently correlated with delayed recurrence or delayed death in multivariate analysis and was associated with increased expression of interferon-gamma, interleukin-2, and lymphocyte-attracting chemokines within the tumor. The absence of intratumoral T cells was associated with increased levels of vascular endothelial growth factor. CONCLUSIONS The presence of intratumoral T cells correlates with improved clinical outcome in advanced ovarian carcinoma.
Collapse
Affiliation(s)
- Lin Zhang
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Silver DF, Hempling RE, Piver MS, Repasky EA. Flt-3 ligand inhibits growth of human ovarian tumors engrafted in severe combined immunodeficient mice. Gynecol Oncol 2000; 77:377-82. [PMID: 10831345 DOI: 10.1006/gyno.2000.5782] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The current study evaluated the effects of Flt-3 ligand (FL) on the growth of human malignant ovarian tumors engrafted in severe combined immunodeficient (SCID) mice with particular attention directed at FL's effect on the host natural killer (NK) cell response against ovarian cancer xenografts. METHODS Equal portions of surgical specimen-derived human ovarian carcinomas were engrafted subcutaneously (SC) into SCID mice. Mice were placed into one of two treatment groups 7 days after the day of implantation. Group 1 received placebo injections SC from Day 1 to Day 20 and group 2 received FL at 10 microg/day SC from Day 1 to Day 20. NK cell depletion was performed on three additional mice from group 2 starting on Day 0 using anti-asialo GM1. Serial tumor volumes were measured. On Day 21, mice from each group were sacrificed, and tumors and spleens were evaluated. Data analysis included chi(2) tests, Student t tests, and analyses of variance when appropriate. RESULTS FL resulted in tumor growth delay compared with control (P = 0.036). When NK cell activity was depleted prior to FL administration, no tumor growth delay was observed. Spleens from FL-treated mice were larger (P < 0.01) with expanded white pulp compared with controls. Histologic examination of tumor sections from FL-treated mice revealed regions of solid tumor growth with glandular architecture similar to that seen in control tumors; however, there was an obvious increase in regions composed largely of dense fibrosis in the FL-treated tumors. NK cells and other infiltrating cells could be detected in clusters among tumors from mice treated with FL whereas these cells were only occasionally detected in sections of control tumors. CONCLUSION FL treatment resulted in an antitumor response against human ovarian cancer engrafted in SCID mice and this inhibition appears to be largely host NK cell mediated. The tumor inhibition seen in this model is similar to that previously seen using syngeneic tumors grown in an immunocompetent animal model. Results from this model can potentially be extrapolated to treatment of human ovarian cancer patients.
Collapse
Affiliation(s)
- D F Silver
- Division of Gynecologic Oncology, Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | | | | | | |
Collapse
|
29
|
Knutson KL, Schiffman K, Rinn K, Disis ML. Immunotherapeutic approaches for the treatment of breast cancer. J Mammary Gland Biol Neoplasia 1999; 4:353-65. [PMID: 10705919 DOI: 10.1023/a:1018714300217] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The application of immunotherapeutic principles to the treatment and prevention of breast cancer is a relatively new undertaking. Although cytokine infusions, cancer vaccines, and T cell therapy have been extensively studied in solid tumors such as melanoma and renal cell carcinoma, the therapeutic efficacy of these approaches is not well explored in breast cancer. The recent definition of tumor-specific immunity in breast cancer patients and the identification of several breast cancer antigens has generated enthusiasm for the application of immune-based therapies to the treatment of breast malignancies. In general, immunotherapies can be considered either non-specific, such as a general immunomodulator (e.g., a cytokine), or tumor-specific (e.g., a vaccine that targets breast cancer tumor antigens). This review describes three major immunotherapeutic strategies that have the potential to enhance or generate an anti-breast cancer T cell immune response: (i) cytokine therapy; (ii) cancer vaccines; and (iii) T cell therapy, and explores how each strategy has been applied to the treatment of breast cancer.
Collapse
Affiliation(s)
- K L Knutson
- Division of Oncology, University of Washington, Seattle 98195-6527, USA
| | | | | | | |
Collapse
|
30
|
Katsetos CD, Stadnicka I, Boyd JC, Ehya H, Zheng S, Soprano CM, Cooper HS, Patchefsky AS, Soprano DR, Soprano KJ. Cellular distribution of retinoic acid receptor-alpha protein in serous adenocarcinomas of ovarian, tubal, and peritoneal origin: comparison with estrogen receptor status. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:469-80. [PMID: 9708807 PMCID: PMC1852976 DOI: 10.1016/s0002-9440(10)65590-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/18/1998] [Indexed: 11/16/2022]
Abstract
Retinoids are effective growth modulators of human ovarian carcinoma cell lines. Their effects are mediated by nuclear retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which are transcriptional factors and members of the steroid/thyroid receptor superfamily. To our knowledge, until now, the cellular distribution of RAR proteins in human ovarian tumor specimens is unknown. This study provides new data on the differential cellular localization of RAR alpha protein in 16 serous adenocarcinomas originating from the ovaries, fallopian tubes, and the peritoneum. Using an affinity-purified antiserum specific for RAR alpha and a monoclonal antibody recognizing the full-length estrogen receptor molecule (clone 6F11), we performed immunohistochemistry on frozen tissue sections and examined the relationship between RAR alpha and estrogen receptor protein expression by comparing the percentage of immunostained tumor cells for either receptor. Our findings indicate a strong linear relationship between the percentages of RAR alpha- and estrogen receptor-labeled tumor cells as determined by linear regression analysis (P < 0.005, r = 0.825). A modest inverse relationship was found between the percentage of RAR alpha-positive tumor cells and histological grade, attesting to a differentiation-dependent trend (P < 0.04). No significant relationship was found between RAR alpha-labeled cells and clinical stage (P = 0.139), site of tumor origin (ovaries versus fallopian tubes versus peritoneum) (P = 0.170), and primary versus metastatic lesion (P = 0.561). Thus, serous adenocarcinomas are capable of expressing RAR alpha and estrogen receptor despite high histological grade and advanced stage of neoplastic disease. Compared with the heterogeneous localization of RAR alpha in cancer cells, there was widespread RAR alpha immunoreactivity in tumor-infiltrating lymphocytes, vascular endothelial cells, and stromal fibroblasts, underscoring the value of immunohistochemistry in the accurate determination of RAR/(RXR) content in tumor specimens.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adult
- Aged
- Aged, 80 and over
- Blotting, Western
- Fallopian Tube Neoplasms/metabolism
- Fallopian Tube Neoplasms/pathology
- Female
- Humans
- Immunoenzyme Techniques
- Middle Aged
- Neoplasms, Cystic, Mucinous, and Serous/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Peritoneal Neoplasms/metabolism
- Peritoneal Neoplasms/pathology
- Receptors, Estrogen/metabolism
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
Collapse
Affiliation(s)
- C D Katsetos
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nash MA, Lenzi R, Edwards CL, Kavanagh JJ, Kudelka AP, Verschraegen CF, Platsoucas CD, Freedman RS. Differential expression of cytokine transcripts in human epithelial ovarian carcinoma by solid tumour specimens, peritoneal exudate cells containing tumour, tumour-infiltrating lymphocyte (TIL)-derived T cell lines and established tumour cell lines. Clin Exp Immunol 1998; 112:172-80. [PMID: 9649178 PMCID: PMC1904977 DOI: 10.1046/j.1365-2249.1998.00576.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
T cell lines derived in low concentrations of recombinant IL-2 (rIL-2) from TIL of patients with epithelial ovarian carcinoma (EOC) often exhibit specific cytotoxicity against autologous tumour cells. However, the ability of T cells at the tumour site to respond to ovarian carcinoma cells may be affected by the production of cytokines by the various cell types present. Using reverse transcriptase-polymerase chain reaction (RT-PCR) we investigated cytokine transcripts in: (i) established EOC tumour cell lines; (ii) solid tumour specimens or peritoneal exudate cells (PEC) from ascites or peritoneal washings of patients with EOC; and (iii) CD4+ TCRalphabeta+ and CD8+ TCRalphabeta+ TIL-derived T cell lines developed in rIL-2. We have found that (i) established EOC tumour cell lines expressed transcripts for transforming growth factor-beta 2 (TGF-beta2) (7/7), but not IL-10 (0/7) or interferon-gamma (IFN-gamma) (0/7) and rarely IL-2 (1/7); (ii) PEC expressed transcripts for IL-2 (12/13), IL-10 (9/13), and TGF-beta2 (12/13), and less often, IFN-gamma (3/13), whereas solid tumour specimens from eight patients with EOC expressed transcripts for IL-2 (4/8), TGF-beta2 (4/8), and IL-10 (5/8), but not for IFN-gamma (0/8); (iii) CD4+ TCRalphabeta+ T cell lines expressed transcripts for IFN-gamma (4/4), IL-2 (4/4) and IL-10 (3/4), whereas CD8+ TCRalphabeta+ T cell lines expressed transcripts for IFN-gamma (5/5), IL-2 (1/5) and IL-10 (2/5). None of these T cell lines expressed TGF-beta2 transcripts. The frequency of IL-2 and TGF-beta2 transcripts in solid tumours was significantly lower than in the PEC (P = 0.0475). CD4+ or CD8+ T cell lines expressing IFN-gamma, IL-2 and IL-10 transcripts were derived in culture with rIL-2 from the TIL of specimens that did not necessarily express these cytokines in the absence of rIL-2. The frequency of cytokine transcripts in T cell lines compared with these same transcripts in the PEC was significantly higher for IFN-gamma (P = 0.0005) and lower for TGF-beta2 (P = 0.0001). An association was observed between the expression of cytokine transcripts in vivo or by TIL-derived cell lines and functions exhibited by either production of cytokines or in vitro cytotoxicity.
Collapse
Affiliation(s)
- M A Nash
- Department of Gynecological Oncology, The University of Texas, M. D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|