301
|
Niccolai E, Cappello P, Taddei A, Ricci F, D'Elios MM, Benagiano M, Bechi P, Bencini L, Ringressi MN, Coratti A, Cianchi F, Bonello L, Di Celle PF, Prisco D, Novelli F, Amedei A. Peripheral ENO1-specific T cells mirror the intratumoral immune response and their presence is a potential prognostic factor for pancreatic adenocarcinoma. Int J Oncol 2016; 49:393-401. [PMID: 27210467 DOI: 10.3892/ijo.2016.3524] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/29/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with an average survival of 4-6 months following diagnosis. Surgical resection is the only treatment with curative intent, but resectable PDAC patients are in the minority. Also, unlike other neoplasms, PDAC is resistant to conventional and targeted chemotherapy. Innovative treatments, such as immunotherapy, can be very important and the study of the immune response is fundamental. We previously demonstrated that PDAC patients show tumor-infiltrating T cells specific to α-enolase (ENO1), a glycolytic enzyme over-expressed by pancreatic tumor cells, which plays an important role in promoting cell migration and cancer metastasis. In the present study, we evaluate the functional anticancer proprieties of ENO1-specific T cells isolated from the peripheral blood of PDAC patients. Furthermore, comparing the T cell receptor repertoire of ENO1-specific peripheral and infiltrating tumor T cells from the same patient suggests that ENO1-specific T cells, despite having a different functional profile, can recirculate from the tumor to the periphery. Finally, of clinical relevance, the presence of peripheral ENO1-specific T cells has a prognostic value and significantly correlates with a longer survival.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Paola Cappello
- Centre for Experimental Research and Medical Studies (CERMS), AOU City of Health and Science of Turin, and Department of Molecular Biotechnology and Health Sciences, University of Turin, I-10126 Turin, Italy
| | - Antonio Taddei
- Department of Surgery and Translational Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Federica Ricci
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Paolo Bechi
- Department of Surgery and Translational Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Lapo Bencini
- Division of General and Oncologic Surgery, Department of Oncology, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Maria Novella Ringressi
- Department of Surgery and Translational Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Andrea Coratti
- Division of General and Oncologic Surgery, Department of Oncology, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Fabio Cianchi
- Department of Surgery and Translational Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Lisa Bonello
- Centre for Experimental Research and Medical Studies (CERMS), AOU City of Health and Science of Turin, and Department of Molecular Biotechnology and Health Sciences, University of Turin, I-10126 Turin, Italy
| | - Paola Francia Di Celle
- General Anatomopathology and Molecular Oncogenetics - AOU City of Health and Science of Turin, I-10126 Turin, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| | - Francesco Novelli
- Centre for Experimental Research and Medical Studies (CERMS), AOU City of Health and Science of Turin, and Department of Molecular Biotechnology and Health Sciences, University of Turin, I-10126 Turin, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, and Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), I-50134 Florence, Italy
| |
Collapse
|
302
|
The SDF-1 rs1801157 Polymorphism is Associated with Cancer Risk: An Update Pooled Analysis and FPRP Test of 17,876 Participants. Sci Rep 2016; 6:27466. [PMID: 27265091 PMCID: PMC4893747 DOI: 10.1038/srep27466] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/17/2016] [Indexed: 02/05/2023] Open
Abstract
The stromal cell derived factor-1 (SDF-1) rs1801157 gene polymorphism has been implicated in susceptibility to cancer, but the results were inconclusive. The current study was to precisely investigate the association between SDF-1 rs1801157 polymorphism and cancer risk using meta-analysis and the false positive report probability (FPRP) test. All 17,876 participants were included in the study. The meta-analysis results indicated a significant association between the SDF-1 rs1801157 polymorphism and cancer risk. By subgroup analyses, the results detected that the SDF-1 rs1801157 polymorphism was associated with cancer susceptibility among Asians and Caucasians. Additionally, we also found significant associations between the SDF-1 rs1801157 polymorphism and susceptibility to different types of cancer. However, to avoid a "false positive report", we further investigated the significant associations observed in the present meta-analysis using the FPRP test. Interestingly, the results of the FPRP test indicated that only 4 gene models were truly associated with cancer risk, especially in Asians. Moreover, we confirmed that the SDF-1 rs1801157 gene polymorphism was only associated with lung and urologic cancer risk. In summary, this study suggested that the SDF-1 rs1801157 polymorphism may serve as a risk factor for cancer development among Asians, especially an increased risk of urologic and lung cancers.
Collapse
|
303
|
Mego M, Gao H, Cohen EN, Anfossi S, Giordano A, Sanda T, Fouad TM, De Giorgi U, Giuliano M, Woodward WA, Alvarez RH, Valero V, Ueno NT, Hortobagyi GN, Cristofanilli M, Reuben JM. Circulating Tumor Cells (CTC) Are Associated with Defects in Adaptive Immunity in Patients with Inflammatory Breast Cancer. J Cancer 2016; 7:1095-104. [PMID: 27326253 PMCID: PMC4911877 DOI: 10.7150/jca.13098] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/07/2015] [Indexed: 01/15/2023] Open
Abstract
Background: Circulating tumor cells (CTCs) play a crucial role in tumor dissemination and are prognostic in primary and metastatic breast cancer. Peripheral blood (PB) immune cells contribute to an unfavorable microenvironment for CTC survival. This study aimed to correlate CTCs with the PB T-cell immunophenotypes and functions of patients with inflammatory breast cancer (IBC). Methods: This study included 65 IBC patients treated at the MD Anderson Cancer Center. PB was obtained from patients prior to starting a new line of chemotherapy for CTCs enumeration by CellSearch®, and T cell phenotype and function by flow cytometry; the results were correlated with CTCs and clinical outcome. Results: At least 1 CTC (≥1) or ≥5 CTCs was detected in 61.5% or 32.3% of patients, respectively. CTC count did not correlate with total lymphocytes; however, patients with ≥1 CTC or ≥5 CTCs had lower percentages (%) of CD3+ and CD4+ T cells compared with patients with no CTCs or <5 CTCs, respectively. Patients with ≥1 CTC had a lower percentage of T-cell receptor (TCR)-activated CD8+ T cells synthesizing TNF-α and IFN-γ and a higher percentage of T-regulatory lymphocytes compared to patients without CTCs. In multivariate analysis, tumor grade and % CD3+ T-cells were associated with ≥1 CTC, whereas ≥5 CTC was associated with tumor grade, stage, % CD3+ and % CD4+ T cells, and % TCR-activated CD8 T-cells synthesizing IL-17. Conclusions: IBC patients with CTCs in PB had abnormalities in adaptive immunity that could potentially impact tumor cell dissemination and initiation of the metastatic cascade.
Collapse
Affiliation(s)
- M Mego
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 5. Currently at 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - H Gao
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - E N Cohen
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - S Anfossi
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - A Giordano
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - T Sanda
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - T M Fouad
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - U De Giorgi
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 6. Currently at Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) - IRCCS, Meldola (FC), Italy
| | - M Giuliano
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 7. Currently at Department of Clinical Medicine and Surgery, University Federico II, Naples. Italy
| | - W A Woodward
- 3. Department of Radiation Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - R H Alvarez
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 4. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 8. Currently at Cancer Treatment Centers of America, Newnan, GA, USA
| | - V Valero
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 4. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - N T Ueno
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 4. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - G N Hortobagyi
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - M Cristofanilli
- 2. Department of Breast Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 9. Currently at Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA, USA
| | - J M Reuben
- 1. Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA;; 4. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
304
|
Huang Y, Ma C, Zhang Q, Ye J, Wang F, Zhang Y, Hunborg P, Varvares MA, Hoft DF, Hsueh EC, Peng G. CD4+ and CD8+ T cells have opposing roles in breast cancer progression and outcome. Oncotarget 2016; 6:17462-78. [PMID: 25968569 PMCID: PMC4627321 DOI: 10.18632/oncotarget.3958] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022] Open
Abstract
The Cancer Immunoediting concept has provided critical insights suggesting dual functions of immune system during the cancer initiation and development. However, the dynamics and roles of CD4+ and CD8+ T cells in the pathogenesis of breast cancer remain unclear. Here we utilized two murine breast cancer models (4T1 and E0771) and demonstrated that both CD4+ and CD8+ T cells were increased and involved in immune responses, but with distinct dynamic trends in breast cancer development. In addition to cell number increases, CD4+ T cells changed their dominant subsets from Th1 in the early stages to Treg and Th17 cells in the late stages of the cancer progression. We also analyzed CD4+ and CD8+ T cell infiltration in primary breast cancer tissues from cancer patients. We observed that CD8+ T cells are the key effector cell population mediating effective anti-tumor immunity resulting in better clinical outcomes. In contrast, intra-tumoral CD4+ T cells have negative prognostic effects on breast cancer patient outcomes. These studies indicate that CD4+ and CD8+ T cells have opposing roles in breast cancer progression and outcomes, which provides new insights relevant for the development of effective cancer immunotherapeutic approaches.
Collapse
Affiliation(s)
- Yi Huang
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Chunling Ma
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Department of Laboratory Medicine, Women and Children's Health Care Hospital of Linyi City, Linyi, P. R. China.,Molecular Biology Experimental Center, Shandong Medical College, Linyi, P. R. China
| | - Qunyuan Zhang
- Department of Genetics, Washington University School of Medicine in St. Louis, Saint Louis, MO, USA
| | - Jian Ye
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Fang Wang
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Yanping Zhang
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Pamela Hunborg
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Mark A Varvares
- Department of Otolaryngology-Head and Neck Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Daniel F Hoft
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Eddy C Hsueh
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Guangyong Peng
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
305
|
Bhadurihauck A, Li L, Li Q, Wang J, Xiao Z. Transient exposure to proteins SOX2, Oct-4, and NANOG immortalizes exhausted tumor-infiltrating CTLs. Biochem Biophys Res Commun 2016; 473:1255-1260. [PMID: 27084449 DOI: 10.1016/j.bbrc.2016.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022]
Abstract
Adoptive cell transfer therapy (ACT) is one of the most promising immunotherapies against cancer, using tumor-infiltrating lymphocytes (TILs) expanded in vitro. Tumor-infiltrating cytotoxic T lymphocytes (TICTLs) play a prominent role in cancer control. TILs terminally differentiate in response to immunosuppressive environments within tumors, and thus are slow to expand and challenging to maintain both in vitro and in patients. To reverse this exhaustion, we utilize a nuclear protein delivery system that exposes TICTLs to the SOX2, Oct-4, and NANOG (SON) proteins. Unlike activated naïve CTLs (effector CTLs), TICTLs respond favorably to SON treatment, exhibiting steady proliferation and extended survivability independent of cytokine and antigen stimulation. Though TICTLs treated with SON (STICTLs) still express T cell receptors as well as other critical downstream components, they are unresponsive to antigen challenge, suggesting that SON treatment regresses TICTLs into a state similar to that of an early double negative T cell. Our findings indicate the TICTL response to SON proteins is unique when compared to effector CTLs, suggesting TICTLs may be sensitive to regulation by other lineage-specific transcription factors and opening a promising new avenue into cancer immunotherapy. To our knowledge, this is the first report on lineage reprogramming of TILs using protein stem cell transcription factors delivered directly to the nucleus.
Collapse
Affiliation(s)
- Anjuli Bhadurihauck
- Department of Animal and Avian Sciences, University of Maryland, College Park, 20742, MD, USA
| | - Lei Li
- Department of Animal and Avian Sciences, University of Maryland, College Park, 20742, MD, USA
| | - Qianqian Li
- Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, 48201, USA
| | - Jianjun Wang
- Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, 48201, USA
| | - Zhengguo Xiao
- Department of Animal and Avian Sciences, University of Maryland, College Park, 20742, MD, USA.
| |
Collapse
|
306
|
Nanda R, Chow LQM, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Pathiraja K, Aktan G, Cheng JD, Karantza V, Buisseret L. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J Clin Oncol 2016; 34:2460-7. [PMID: 27138582 DOI: 10.1200/jco.2015.64.8931] [Citation(s) in RCA: 1137] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Immune checkpoint inhibition has been demonstrated to be an effective anticancer strategy. Several lines of evidence support the study of immunotherapy in triple-negative breast cancer (TNBC). We assessed the safety and antitumor activity of the programmed cell death protein 1 (PD-1) inhibitor pembrolizumab in patients with advanced TNBC. METHODS KEYNOTE-012 (ClinicalTrials.gov identifier: NCT01848834) was a multicenter, nonrandomized phase Ib trial of single-agent pembrolizumab given intravenously at 10 mg/kg every 2 weeks to patients with advanced PD-L1-positive (expression in stroma or ≥ 1% of tumor cells by immunohistochemistry) TNBC, gastric cancer, urothelial cancer, and head and neck cancer. This report focuses on the TNBC cohort. RESULTS Among 111 patients with TNBC whose tumor samples were screened for PD-L1 expression, 58.6% had PD-L1-positive tumors. Thirty-two women (median age, 50.5 years; range, 29 to 72 years) were enrolled and assessed for safety and antitumor activity. The median number of doses administered was five (range, 1 to 36 doses). Common toxicities were mild and similar to those observed in other tumor cohorts (eg, arthralgia, fatigue, myalgia, and nausea), and included five (15.6%) patients with grade ≥ 3 toxicity and one treatment-related death. Among the 27 patients who were evaluable for antitumor activity, the overall response rate was 18.5%, the median time to response was 17.9 weeks (range, 7.3 to 32.4 weeks), and the median duration of response was not yet reached (range, 15.0 to ≥ 47.3 weeks). CONCLUSION This phase Ib study describes preliminary evidence of clinical activity and a potentially acceptable safety profile of pembrolizumab given every 2 weeks to patients with heavily pretreated, advanced TNBC. A single-agent phase II study examining a 200-mg dose given once every 3 weeks (ClinicalTrials.gov identifier: NCT02447003) is ongoing.
Collapse
Affiliation(s)
- Rita Nanda
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium.
| | - Laura Q M Chow
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - E Claire Dees
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Raanan Berger
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Shilpa Gupta
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Ravit Geva
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Lajos Pusztai
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Kumudu Pathiraja
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Gursel Aktan
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Jonathan D Cheng
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Vassiliki Karantza
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Laurence Buisseret
- Rita Nanda, University of Chicago, Chicago, IL; Laura Q.M. Chow, University of Washington, Seattle, WA; E. Claire Dees, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Raanan Berger, Sheba Medical Center, Tel Hashomer; Ravit Geva, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Shilpa Gupta, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Lajos Pusztai, Yale University School of Medicine, New Haven, CT; Kumudu Pathiraja, Gursel Aktan, Jonathan D. Cheng, and Vassiliki Karantza, Merck & Co., Kenilworth, NJ; and Laurence Buisseret, Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
307
|
Abstract
Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States.
| |
Collapse
|
308
|
Wiedemann GM, Knott MML, Vetter VK, Rapp M, Haubner S, Fesseler J, Kühnemuth B, Layritz P, Thaler R, Kruger S, Ormanns S, Mayr D, Endres S, Anz D. Cancer cell-derived IL-1α induces CCL22 and the recruitment of regulatory T cells. Oncoimmunology 2016; 5:e1175794. [PMID: 27757295 DOI: 10.1080/2162402x.2016.1175794] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/29/2016] [Accepted: 04/04/2016] [Indexed: 01/04/2023] Open
Abstract
In cancer patients, immunosuppression through regulatory T cells (Treg) is a crucial component of tumor immune evasion and contributes to disease progression. Tumor-infiltrating Treg in particular suppress local effector T cell responses and are associated with poor prognosis in tumors such as human pancreatic cancer or hepatocellular carcinoma (HCC). The chemokine CCL22 is known to recruit Treg into the tumor tissue and many types of human tumors are known to express high levels of CCL22. The mechanisms leading to intratumoral secretion of CCL22 are so far unknown. We demonstrate here that intratumoral CCL22 is induced in tumor-infiltrating immune cells through cancer cell-derived interleukin-1 (IL-1α). In pancreatic cancer and HCC, CCL22 is produced by intratumoral dendritic cells, while the cancer cells themselves do not secrete CCL22 in vitro and in vivo. Incubation of human peripheral blood mononuclear cells (PBMC) or murine splenocytes with tumor cells or tumor cell supernatants strongly induced CCL22 secretion in vitro. Tumor cell supernatants contained IL-1 and CCL22 induction in PBMC could be specifically prevented by the IL-1 receptor antagonist anakinra or by transfection of tumor cell lines with IL-1 siRNA, leading to a suppression of Treg migration. In conclusion, we identify here tumor cell-derived IL-1α as a major inducer of the Treg attracting chemokine CCL22 in human cancer cells. Therapeutic blockade of the IL-1 pathway could represent a promising strategy to inhibit tumor-induced immunosuppression.
Collapse
Affiliation(s)
- Gabriela Maria Wiedemann
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Max Martin Ludwig Knott
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Viola Katharina Vetter
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Moritz Rapp
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Sascha Haubner
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Julia Fesseler
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Benjamin Kühnemuth
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Patrick Layritz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Raffael Thaler
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München , Munich, Germany
| | - Stephan Kruger
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München , Munich, Germany
| | - Steffen Ormanns
- Department of Pathology, Ludwig-Maximilians-Universität , Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians-Universität , Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
309
|
Brand JS, Colzani E, Johansson ALV, Giesecke J, Clements M, Bergh J, Hall P, Czene K. Infection-related hospitalizations in breast cancer patients: Risk and impact on prognosis. J Infect 2016; 72:650-658. [PMID: 27063280 DOI: 10.1016/j.jinf.2016.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/04/2016] [Accepted: 04/03/2016] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Infections are a common cause of hospitalization in breast cancer patients. We studied the risk, clinical characteristics and outcomes of infection-related hospitalizations in this patient population. METHODS A Swedish registry-based study including 8338 breast cancer patients diagnosed between 2001 and 2008, followed prospectively for infection-related hospitalizations until 2010. Standardized incidence ratios (SIRs) were calculated using background rates from the general female population. Associations with clinical characteristics and mortality were analyzed using flexible parametric survival models. RESULTS In total, 720 patients experienced an infection-related hospitalization during a median follow-up of 4.9 years. Infection rates were highest within the first year of diagnosis (SIR = 5.61, 95% CI; 4.98-6.32), and site-specific risks were most pronounced for sepsis (SIR = 3.14, 95% CI; 2.66-3.71) and skin infections (SIR = 2.80, 95% CI; 2.24-3.50). Older age at diagnosis, comorbidities, markers of tumor aggressiveness, chemotherapy and axillary node dissection were independent predictors of infectious disease risk. Infection-related hospitalizations were also independently associated with overall and breast cancer-specific death. CONCLUSIONS A significant number of breast cancer patients are hospitalized with an infection following diagnosis, which in turn predicts poor prognosis. The risk profile of infection-related hospitalizations is multifactorial, including patient, tumor and treatment-related factors.
Collapse
Affiliation(s)
- Judith S Brand
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Edoardo Colzani
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anna L V Johansson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Giesecke
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mark Clements
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Radiumhemmet, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
310
|
Adenosine-mediated immunosuppression in patients with squamous cell carcinoma of the head and neck. HNO 2016; 64:303-9. [DOI: 10.1007/s00106-016-0137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
311
|
Zhang F, Dong W, Zeng W, Zhang L, Zhang C, Qiu Y, Wang L, Yin X, Zhang C, Liang W. Naringenin prevents TGF-β1 secretion from breast cancer and suppresses pulmonary metastasis by inhibiting PKC activation. Breast Cancer Res 2016; 18:38. [PMID: 27036297 PMCID: PMC4818388 DOI: 10.1186/s13058-016-0698-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/15/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Targeting the TGF-β1 pathway for breast cancer metastasis therapy has become an attractive strategy. We have previously demonstrated that naringenin significantly reduced TGF-β1 levels in bleomycin-induced lung fibrosis and effectively prevented pulmonary metastases of tumors. This raised the question of whether naringenin can block TGF-β1 secretion from breast cancer cells and inhibit their pulmonary metastasis. METHODS We transduced a lentiviral vector encoding the mouse Tgf-β1 gene into mouse breast carcinoma (4T1-Luc2) cells and inoculated the transformant cells (4T1/TGF-β1) into the fourth primary fat pat of Balb/c mice. Pulmonary metastases derived from the primary tumors were monitored using bioluminescent imaging. Spleens, lungs and serum (n = 18-20 per treatment group) were analyzed for immune cell activity and TGF-β1 level. The mechanism whereby naringenin decreases TGF-β1 secretion from breast cancer cells was investigated at different levels, including Tgf-β1 transcription, mRNA stability, translation, and extracellular release. RESULTS In contrast to the null-vector control (4T1/RFP) tumors, extensive pulmonary metastases derived from 4T1/TGF-β1 tumors were observed. Administration of the TGF-β1 blocking antibody 1D11 or naringenin showed an inhibition of pulmonary metastasis for both 4T1/TGF-β1 tumors and 4T1/RFP tumors, resulting in increased survival of the mice. Compared with 4T1/RFP bearing mice, systemic immunosuppression in 4T1/TGF-β1 bearing mice was observed, represented by a higher proportion of regulatory T cells and myeloid-derived suppressor cells and a lower proportion of activated T cells and INFγ expression in CD8(+) T cells. These metrics were improved by administration of 1D11 or naringenin. However, compared with 1D11, which neutralized secreted TGF-β1 but did not affect intracellular TGF-β1 levels, naringenin reduced the secretion of TGF-β1 from the cells, leading to an accumulation of intracellular TGF-β1. Further experiments revealed that naringenin had no effect on Tgf-β1 transcription, mRNA decay or protein translation, but prevented TGF-β1 transport from the trans-Golgi network by inhibiting PKC activity. CONCLUSIONS Naringenin blocks TGF-β1 trafficking from the trans-Golgi network by suppressing PKC activity, resulting in a reduction of TGF-β1 secretion from breast cancer cells. This finding suggests that naringenin may be an attractive therapeutic candidate for TGF-β1 related diseases.
Collapse
Affiliation(s)
- Fayun Zhang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenjuan Dong
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenfeng Zeng
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Zhang
- Department of Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434000, China
| | - Chao Zhang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuqi Qiu
- Department of Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434000, China
| | - Luoyang Wang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaozhe Yin
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chunling Zhang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
312
|
Liao W, Wang Y, Liao Y, He S, Jin J. Preoperative Aspartate Aminotransferase to White Blood Cell Count Ratio Predicting Postoperative Outcomes of Hepatocellular Carcinoma. Medicine (Baltimore) 2016; 95:e3345. [PMID: 27057915 PMCID: PMC4998831 DOI: 10.1097/md.0000000000003345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Effective biomarkers for predicting prognosis of hepatocellular carcinoma (HCC) patients after hepatectomy is urgently needed. The purpose of this study is to evaluate the value of the preoperative peripheral aspartate aminotransferase to white blood cell count ratio (AWR) for the prognostication of patients with HCC.Clinical data of 396 HCC patients who underwent radical hepatectomy were retrospectively analyzed. The patients were divided into the low-AWR group (AWR ≤5.2) and the high-AWR group (AWR >5.2); univariate analysis, Kaplan-Meier method analysis, and the multivariate analysis by Cox regression were conducted, respectively.The results showed that AWR was associated with alpha-fetoprotein (AFP), tumor size, Barcelona clinic liver cancer (BCLC) stage, portal vein tumor thrombus (PVTT), and alanine aminotransferase (ALT) in HCC. AWR > 5.2, AFP > 100 ng/mL, size of tumor >6 cm, number of multiple tumors, B-C of BCLC stage, PVTT, and distant metastasis were predictors of poorer disease-free survival (DFS) and overall survival (OS). Except for recurrence, which was an independent predictor for OS only, AWR >5.2, size of tumor >6 cm, and PVTT were independent predictors of both DFS and OS.We concluded that preoperative AWR > 5.2 was an adverse predictor of DFS and OS in HCC after hepatectomy, AWR might be a novel prognostic biomarker in HCC after curative resection.
Collapse
Affiliation(s)
- Weijia Liao
- From the Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University (WL, YW, SH, JJ); Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair (WL, YW, SH, JJ); Disease Prevention and Control Center of Guilin (YL); and China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi, China (JJ)
| | | | | | | | | |
Collapse
|
313
|
Partecke LI, Speerforck S, Käding A, Seubert F, Kühn S, Lorenz E, Schwandke S, Sendler M, Keßler W, Trung DN, Oswald S, Weiss FU, Mayerle J, Henkel C, Menges P, Beyer K, Lerch MM, Heidecke CD, von Bernstorff W. Chronic stress increases experimental pancreatic cancer growth, reduces survival and can be antagonised by beta-adrenergic receptor blockade. Pancreatology 2016; 16:423-33. [PMID: 27083074 DOI: 10.1016/j.pan.2016.03.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 02/17/2016] [Accepted: 03/06/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Chronic stress could promote tumour growth and reduce survival of pancreatic cancer patients via beta-adrenergic receptors of tumour cells. We have tested the impact of chronic acoustic and restraint stress on tumour development in an orthotopic syngeneic murine model of pancreatic cancer. METHODS AND RESULTS Tumour-bearing C57BL/6 mice exposed to chronic stress had 45% (p = 0.0138) higher circulating steroid and 111% (p = 0.0052) higher adrenal tyrosine hydroxylase levels. Their immune response was significantly suppressed: The in vitro LPS response of splenocytes was significantly reduced regarding Th1- and Th2-cytokines including IFN-gamma, IL-6, IL-10 and MCP-1 (0.0011 < p < 0.043). Also, tumours of stressed mice showed a tendency towards fewer total CD4 cells, more regulatory T cells (Treg), less T cell/tumour cell contacts and a reduction of CTLA-4 in CD4 cells (p > 0.05). TGF-beta in vitro was increased by 23.4% using catecholamines (p < 0.012) and in vivo employing chronic stress (p < 0.001). After 5 weeks tumour volumes were 130% (p = 0.0061) larger and median survival reduced by 13.5% (p = 0.0058). Tumours expressed more VEGF (p = 0.0334), had greater microvessel densities (p = 0.047), and an increased MMP-9 expression (p = 0.0456). Beta-catecholamines increased proliferation in tumour cells by 18% (p < 0.0001) and migration by 78% (p = 0.0348) whereas the beta-blocker propranolol reduced these effects by 25% (p < 0.0001) and 53% (p = 0.045), respectively. When stressed tumour-bearing animals were treated with propranolol tumour volumes were reduced by 69% (p = 0.0088) and survival improved by 14% (p < 0.0058). CONCLUSIONS The potential treatment with beta-blockers of patients with pancreatic cancer or other malignancies should be further evaluated as an adjuvant anti-neoplastic agent in clinical trials.
Collapse
Affiliation(s)
- Lars Ivo Partecke
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Sven Speerforck
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - André Käding
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Florian Seubert
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Sandra Kühn
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Eric Lorenz
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Sebastian Schwandke
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Wolfram Keßler
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Dung Nguyen Trung
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Stefan Oswald
- Institute of Pharmacology, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Christin Henkel
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Pia Menges
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Katharina Beyer
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Claus-Dieter Heidecke
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Wolfram von Bernstorff
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany.
| |
Collapse
|
314
|
Hickman ES, Lomax ME, Jakobsen BK. Antigen Selection for Enhanced Affinity T-Cell Receptor-Based Cancer Therapies. ACTA ACUST UNITED AC 2016; 21:769-85. [PMID: 26993321 DOI: 10.1177/1087057116637837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/15/2016] [Indexed: 12/11/2022]
Abstract
Evidence of adaptive immune responses in the prevention of cancer has been accumulating for decades. Spontaneous T-cell responses occur in multiple indications, bringing the study of de novo expressed cancer antigens to the fore and highlighting their potential as targets for cancer immunotherapy. Circumventing the immune-suppressive mechanisms that maintain tumor tolerance and driving an antitumor cytotoxic T-cell response in cancer patients may eradicate the tumor or block disease progression. Multiple strategies are being pursued to harness the cytotoxic potential of T cells clinically. Highly promising results are now emerging. The focus of this review is the target discovery process for cancer immune therapeutics based on affinity-matured T-cell receptors (TCRs). Target cancer antigens in the context of adoptive cell transfer technologies and soluble biologic agents are discussed. To appreciate the impact of TCR-based technology and understand the TCR discovery process, it is necessary to understand key differences between TCR-based therapy and other immunotherapy approaches. The review first summarizes key advances in the cancer immunotherapy field and then discusses the opportunities that TCR technology provides. The nature and breadth of molecular targets that are tractable to this approach are discussed, together with the challenges associated with finding them.
Collapse
|
315
|
Chen X, Du Y, Lin X, Qian Y, Zhou T, Huang Z. CD4+CD25+ regulatory T cells in tumor immunity. Int Immunopharmacol 2016; 34:244-249. [PMID: 26994448 DOI: 10.1016/j.intimp.2016.03.009] [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] [Received: 01/14/2016] [Revised: 02/22/2016] [Accepted: 03/08/2016] [Indexed: 01/11/2023]
Abstract
Regulatory T cells (Tregs) are essential for maintaining peripheral tolerance, preventing autoimmune diseases and limiting chronic inflammatory diseases. Depletion of Tregs results in the onset of a variety of autoimmune diseases. Tregs are defined based on expression of CD4, CD25, and the transcription factor, FoxP3. It is now clear that three inhibitory cytokines, IL-10, IL-35 and TGF-β, are key mediators of Tregs function. Tregs have been shown to be important contributors to the development of immune tolerance toward tumors and play a critical role in the induction of tolerance to tumor associated antigens and suppression of anti-tumor immunity. Increasing researches support the existence of elevated numbers of regulatory T cells in cancer patients. Poor prognosis and decreased survival rates are closely correlated with higher Treg cell frequencies. Depletion of Tregs or blockade of their immune inhibitory role can enhance anti-tumor effects. Recent evidence suggests that Tregs may be responsible for the failure of host anti-tumor immunity by suppressing cytotoxic T-cells. In this review, we discuss cellular and molecular mechanisms in the differentiation and function of Tregs in tumor immunity.
Collapse
Affiliation(s)
- Xin Chen
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yong Du
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - XiuQing Lin
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yan Qian
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Ting Zhou
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - ZhiMing Huang
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
316
|
Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors. SCIENCE CHINA-LIFE SCIENCES 2016; 59:340-8. [PMID: 26965525 DOI: 10.1007/s11427-016-5027-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/19/2016] [Indexed: 12/20/2022]
|
317
|
Halvorsen EC, Hamilton MJ, Young A, Wadsworth BJ, LePard NE, Lee HN, Firmino N, Collier JL, Bennewith KL. Maraviroc decreases CCL8-mediated migration of CCR5(+) regulatory T cells and reduces metastatic tumor growth in the lungs. Oncoimmunology 2016; 5:e1150398. [PMID: 27471618 DOI: 10.1080/2162402x.2016.1150398] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/21/2016] [Accepted: 01/31/2016] [Indexed: 02/08/2023] Open
Abstract
Regulatory T cells (Tregs) play a crucial physiological role in the regulation of immune homeostasis, although recent data suggest Tregs can contribute to primary tumor growth by suppressing antitumor immune responses. Tregs may also influence the development of tumor metastases, although there is a paucity of information regarding the phenotype and function of Tregs in metastatic target organs. Herein, we demonstrate that orthotopically implanted metastatic mammary tumors induce significant Treg accumulation in the lungs, which is a site of mammary tumor metastasis. Tregs in the primary tumor and metastatic lungs express high levels of C-C chemokine receptor type 5 (CCR5) relative to Tregs in the mammary fat pad and lungs of tumor-free mice, and Tregs in the metastatic lungs are enriched for CCR5 expression in comparison to other immune cell populations. We also identify that C-C chemokine ligand 8 (CCL8), an endogenous ligand of CCR5, is produced by F4/80(+) macrophages in the lungs of mice with metastatic primary tumors. Migration of Tregs toward CCL8 ex vivo is reduced in the presence of the CCR5 inhibitor Maraviroc. Importantly, treatment of mice with Maraviroc (MVC) reduces the level of CCR5(+) Tregs and metastatic tumor burden in the lungs. This work provides evidence of a CCL8/CCR5 signaling axis driving Treg recruitment to the lungs of mice bearing metastatic primary tumors, representing a potential therapeutic target to decrease Treg accumulation and metastatic tumor growth.
Collapse
Affiliation(s)
- E C Halvorsen
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - M J Hamilton
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - A Young
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - B J Wadsworth
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - N E LePard
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - H N Lee
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - N Firmino
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - J L Collier
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - K L Bennewith
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
318
|
Nielsen MFB, Mortensen MB, Detlefsen S. Key players in pancreatic cancer-stroma interaction: Cancer-associated fibroblasts, endothelial and inflammatory cells. World J Gastroenterol 2016; 22:2678-2700. [PMID: 26973408 PMCID: PMC4777992 DOI: 10.3748/wjg.v22.i9.2678] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/19/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is the most aggressive type of common cancers, and in 2014, nearly 40000 patients died from the disease in the United States. Pancreatic ductal adenocarcinoma, which accounts for the majority of PC cases, is characterized by an intense stromal desmoplastic reaction surrounding the cancer cells. Cancer-associated fibroblasts (CAFs) are the main effector cells in the desmoplastic reaction, and pancreatic stellate cells are the most important source of CAFs. However, other important components of the PC stroma are inflammatory cells and endothelial cells. The aim of this review is to describe the complex interplay between PC cells and the cellular and non-cellular components of the tumour stroma. Published data have indicated that the desmoplastic stroma protects PC cells against chemotherapy and radiation therapy and that it might promote the proliferation and migration of PC cells. However, in animal studies, experimental depletion of the desmoplastic stroma and CAFs has led to more aggressive cancers. Hence, the precise role of the tumour stroma in PC remains to be elucidated. However, it is likely that a context-dependent therapeutic modification, rather than pure depletion, of the PC stroma holds potential for the development of new treatment strategies for PC patients.
Collapse
|
319
|
Bhattacharyya S, Saha J. Tumour, Oxidative Stress and Host T Cell Response: Cementing the Dominance. Scand J Immunol 2016; 82:477-88. [PMID: 26286126 DOI: 10.1111/sji.12350] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/10/2015] [Indexed: 12/23/2022]
Abstract
Reactive oxygen species (ROS) and free radicals are produced intrinsically during normal cellular metabolic processes or extrinsically due to ionizing radiations, UV rays, xenobiotic insult, etc. ROS are important signal mediators and are used by the immune system to destroy pathogens, but as these are highly reactive, they also have the capacity to cause DNA damage and alter protein and lipid components of a cell. As a result, cells have evolved a tight regulation of internal redox environment that involves a balanced interplay between free radicals produced and quenched by cellular antioxidants and enzyme systems. Any deregulation of this subtle balance can result in oxidative stress that can lead to various pathological conditions including cancer. Oxidative stress can be a cause of neoplasia, or it can be induced by a growing tumour itself. The link existing between oxidative stress and inflammation is also very strong. Suppressed cellular immune system, especially effector T cell system, is a characteristic of tumour-bearing host. Both the direct oxidative stress caused by tumour cell(s) and oxidative stress mediators present in tumour microenvironment play a significant role in the suppression of effector T cell function and induction of T cell death. This review discusses in detail the complex interplay between tumour-stroma-immune system in the light of oxidative stress that dominates every phase of cancer including initiation, progression and establishment. This review also addresses in detail the mechanisms of oxidative stress-induced T cell dysfunction in tumour-bearing host and also briefly points out the possible therapeutic interventions.
Collapse
Affiliation(s)
- S Bhattacharyya
- Department of Zoology, Sidho Kanho Birsha University, Purulia, West Bengal, India
| | - J Saha
- Department of Zoology, Sidho Kanho Birsha University, Purulia, West Bengal, India
| |
Collapse
|
320
|
Immune Checkpoint Modulators: An Emerging Antiglioma Armamentarium. J Immunol Res 2016; 2016:4683607. [PMID: 26881264 PMCID: PMC4736366 DOI: 10.1155/2016/4683607] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoints have come to the forefront of cancer therapies as a powerful and promising strategy to stimulate antitumor T cell activity. Results from recent preclinical and clinical studies demonstrate how checkpoint inhibition can be utilized to prevent tumor immune evasion and both local and systemic immune suppression. This review encompasses the key immune checkpoints that have been found to play a role in tumorigenesis and, more specifically, gliomagenesis. The review will provide an overview of the existing preclinical and clinical data, antitumor efficacy, and clinical applications for each checkpoint with respect to GBM, as well as a summary of combination therapies with chemotherapy and radiation.
Collapse
|
321
|
Abstract
Flow cytometry is an essential tool for studying the tumor microenvironment. It allows us to quickly quantify and identify multiple cell types in a heterogeneous sample. A brief overview of flow cytometry instrumentation and the appropriate considerations and steps in building a good flow cytometry staining panel are discussed. In addition, a lymphoid tissue and solid tumor leukocyte infiltrate flow cytometry staining protocol and an example of flow cytometry data analysis are presented.
Collapse
Affiliation(s)
- Yoon Kow Young
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Alicia M Bolt
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Ryuhjin Ahn
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Koren K Mann
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada.
| |
Collapse
|
322
|
Sage EK, Schmid TE, Sedelmayr M, Gehrmann M, Geinitz H, Duma MN, Combs SE, Multhoff G. Comparative analysis of the effects of radiotherapy versus radiotherapy after adjuvant chemotherapy on the composition of lymphocyte subpopulations in breast cancer patients. Radiother Oncol 2015; 118:176-80. [PMID: 26683801 DOI: 10.1016/j.radonc.2015.11.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Breast cancer is the most common cancer in women worldwide and surgery, radiotherapy (RT) and chemotherapy (ChT) are frequently used to treat this cancer. Adjuvant RT has been shown to cause long-term changes in lymphocyte counts in the peripheral blood. Herein, the time course of changes in lymphocyte subpopulations upon RT was studied in patients with and without adjuvant ChT in order to explore its potential clinical impact. MATERIALS AND METHODS Total lymphocyte counts and the composition of lymphocyte subpopulations before RT (t0), after 30 Gy (t1), at the end of RT (t2), and 6 weeks (t3), 6 months (t4), and 1 year (t5) after RT were studied by flow cytometry. RESULTS Absolute lymphocyte counts were significantly lower in all breast cancer patients (n=40) before and also 1 year after RT compared to healthy controls. The percentage of CD3(+)/CD4(+) helper T cells and FoxP3(+) regulatory T cells increased significantly in patients without adjuvant ChT. Different NK cell subpopulations dropped during RT in patients with and without ChT, but recovered to initial levels 6months after RT (t4). During RT (t0-t2) the percentage of CD19(+) B cells significantly dropped in patients without ChT, but gradually increased in patients with adjuvant ChT. Both patient groups reached initial levels 6 months after RT (t4). CONCLUSION Different lymphocyte subpopulations respond differently to RT with and without adjuvant ChT. CD4(+) T cells increase during RT, whereas NK cells and B cells decrease in patients without ChT, but recover within 6 months after RT. Treg cells gradually increase in patients without ChT from t0 to t5, but not in patients with adjuvant ChT.
Collapse
Affiliation(s)
- Eva K Sage
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany; HelmholtzZentrum München, Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Germany
| | - Michael Sedelmayr
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany
| | - Mathias Gehrmann
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany
| | - Hans Geinitz
- Department of Radiation Oncology, Krankenhaus der Barmherzigen Schwestern and Medical Faculty, Johannes Kepler University Linz, Austria
| | - Marciana N Duma
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany; HelmholtzZentrum München, Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany; HelmholtzZentrum München, Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Germany; HelmholtzZentrum München, Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Germany.
| |
Collapse
|
323
|
Friedman A, Liao KL. The role of the cytokines IL-27 and IL-35 in cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2015; 12:1203-1217. [PMID: 26775857 DOI: 10.3934/mbe.2015.12.1203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The cancer-immune interaction is a fast growing field of research in biology, where the goal is to harness the immune system to fight cancer more effectively. In the present paper we review recent work of the interaction between T cells and cancer. CD8+ T cells are activated by IL-27 cytokine and they kill tumor cells. Regulatory T cells produce IL-35 which promotes cancer cells by enhancing angiogenesis, and inhibit CD8+ T cells via TGF-β production. Hence injections of IL-27 and anti-IL-35 are both potentially anti-tumor drugs. The models presented here are based on experimental mouse experiments, and their simulations agree with these experiments. The models are used to suggest effective schedules for drug treatment.
Collapse
Affiliation(s)
- Avner Friedman
- Mathematical Biosciences Institute, The Ohio State University, Columbus, OH 43210, United States
| | | |
Collapse
|
324
|
O'Callaghan DS, Rexhepaj E, Gately K, Coate L, Delaney D, O'Donnell DM, Kay E, O'Connell F, Gallagher WM, O'Byrne KJ. Tumour islet Foxp3+ T-cell infiltration predicts poor outcome in nonsmall cell lung cancer. Eur Respir J 2015; 46:1762-72. [DOI: 10.1183/13993003.00176-2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 07/14/2015] [Indexed: 12/29/2022]
Abstract
The impact of host immunity on outcome in nonsmall cell lung cancer (NSCLC) is controversial. We examined the relationship between lymphoid infiltration patterns in NSCLC and prognosis.Tumour- and stroma-infiltrating CD3+, CD8+ and forkhead box P3 (Foxp3)+ T-lymphocytes were identified using immunohistochemistry and a novel image analysis algorithm to assess total, cytotoxic and regulatory T-lymphocyte counts, respectively, in 196 NSCLC cases. The median cell count was selected as a cut-point to define patient subgroups and the ratio of the corresponding tumour islet:stroma (TI/S) counts was determined.There was a positive association between overall survival and increased CD8+ TI/S ratio (hazard ratio (HR) for death 0.44, p<0.001) but an inverse relationship between Foxp3+ TI/S ratio and overall survival (HR 4.86, p<0.001). Patients with high CD8+ islet (HR 0.48, p<0.001) and Foxp3+ stromal (HR 0.23, p<0.001) counts had better survival, whereas high CD3+ and CD8+ stromal counts and high Foxp3+ islet infiltration conferred a worse survival (HR 1.55, 2.19 and 3.14, respectively). By multivariate analysis, a high CD8+ TI/S ratio conferred an improved survival (HR 0.48, p=0.002) but a high Foxp3+ TI/S ratio was associated with worse survival (HR 3.91, p<0.001).Microlocalisation of infiltrating T-lymphocytes is a powerful predictor of outcome in resected NSCLC.
Collapse
|
325
|
Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Arra C, Maiolino P, Izzo F, Tornesello ML, Aurisicchio L, Ciliberto G, Buonaguro FM, Buonaguro L. Novel metronomic chemotherapy and cancer vaccine combinatorial strategy for hepatocellular carcinoma in a mouse model. Cancer Immunol Immunother 2015; 64:1305-1314. [PMID: 25944003 PMCID: PMC11028459 DOI: 10.1007/s00262-015-1698-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/13/2015] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer and represents the third and the fifth leading cause of cancer-related death worldwide in men and women, respectively. Hepatitis B virus (HBV) and hepatitis C virus (HCV) chronic infections account for pathogenesis of more than 80 % of primary HCC. HCC prognosis greatly varies according to stage at beginning of treatment, but the overall 5-year survival rate is approximately 5-6 %. Given the limited number of effective therapeutic strategies available, immunotherapies and therapeutic cancer vaccines may help in improving the clinical outcome for HCC patients. However, the few clinical trials conducted to date have shown contrasting results, indicating the need for improvements. In the present study, a novel combinatorial strategy, based on metronomic chemotherapy plus vaccine, is evaluated in a mouse model. The chemotherapy is a multi-drug cocktail including taxanes and alkylating agents, which is administered in a metronomic-like fashion. The vaccine is a multi-peptide cocktail including HCV as well as universal tumor antigen TERT epitopes. The combinatorial strategy designed and evaluated in the present study induces an enhanced specific T cell response, when compared to vaccine alone, which correlates to a reduced Treg frequency. Such results are highly promising and may pave way to relevant improvements in immunotherapeutic strategies for HCC and beyond.
Collapse
Affiliation(s)
- Maria Tagliamonte
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Annacarmen Petrizzo
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Maria Napolitano
- Laboratory of Clinical Immunology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Francesco Izzo
- Hepato-Biliary Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Maria Lina Tornesello
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | | | - Gennaro Ciliberto
- Scientific Direction, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Franco M. Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| | - Luigi Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” - IRCCS, Naples, Italy
| |
Collapse
|
326
|
Oates J, Hassan NJ, Jakobsen BK. ImmTACs for targeted cancer therapy: Why, what, how, and which. Mol Immunol 2015; 67:67-74. [DOI: 10.1016/j.molimm.2015.01.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 12/20/2022]
|
327
|
Chen X, Zhu B, Luo Y, Zhang D, Zhang L, Zhu H, Hao C, Guo Y, Liu H. Interleukin-28B Plays a Therapeutic Role on Mouse U14 Cervical Cancer Cells by Down-Regulating CD4+CD25+FoxP3+Regulatory T Cells In Vivo. Int J Gynecol Cancer 2015; 25:1369-76. [PMID: 26270121 DOI: 10.1097/igc.0000000000000528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
AIM To investigate the immunotherapeutic effectiveness of adenoviral vector expressing mouse interleukin (IL)-28B (Ad-mIL-28B) against cervical cancer and its mechanism. METHOD U14 cervical cancer cell-bearing mice were treated with Ad-mIL-28B. Meanwhile, whole cell vaccine was prepared by repeated freezing and thawing U14 cells. Then CD4⁺CD25⁺FoxP3⁺regulatory T (Treg) cells were evaluated by flow cytometry. Tumor volume and metastasis in BALB/c and C57BL/6j mice were detected. RESULTS Ad-mIL-28B treatment significantly decreased the number of CD4⁺CD25⁺FoxP3⁺Treg cells. Subsequently, there was a significant decrease in the size of tumor tissue and the numbers of heteromorphic tumor cells. The tumor metastasis in the lung and liver of the Ad-mIL-28B group also decreased. However, there was no therapeutic effect observed for whole cell vaccine on U14 tumor-bearing mice. CONCLUSION Interleukin-28B can inhibit the growth and metastasis of cervical cancer in U14 tumor-bearing mice by down-regulating Treg cells.
Collapse
Affiliation(s)
- Xiaoyun Chen
- *Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Lanzhou University, Lanzhou, Gansu Province, China; Institutes of †Pathogen Biology and ‡Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China; §Institute of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China; and ∥Department of Obstetrics and Gynecology, Gansu Provincial Hospital, Lanzhou, Gansu Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
328
|
Cook AM, McDonnell AM, Lake RA, Nowak AK. Dexamethasone co-medication in cancer patients undergoing chemotherapy causes substantial immunomodulatory effects with implications for chemo-immunotherapy strategies. Oncoimmunology 2015; 5:e1066062. [PMID: 27141331 PMCID: PMC4839331 DOI: 10.1080/2162402x.2015.1066062] [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: 05/11/2015] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 12/29/2022] Open
Abstract
The glucocorticoid (GC) steroid dexamethasone (Dex) is used as a supportive care co-medication for cancer patients undergoing standard care pemetrexed/platinum doublet chemotherapy. As trials for new cancer immunotherapy treatments increase in prevalence, it is important to track the immunological changes induced by co-medications commonly used in the clinic, but not specifically included in trial design or in pre-clinical models. Here, we document a number of Dex -induced immunological effects, including a large-scale lymphodepletive effect particularly affecting CD4+ T cells but also CD8+ T cells. The proportion of regulatory T cells within the CD4+ compartment did not change after Dex was administered, however a significant increase in proliferation and activation of regulatory T cells was observed. We also noted Dex -induced proportional changes in dendritic cell (DC) subtypes. We discuss these immunological effects in the context of chemoimmunotherapy strategies, and suggest a number of considerations to be taken into account when designing future studies where Dex and other GCs may be in use.
Collapse
Affiliation(s)
- Alistair M Cook
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia; National Centre for Asbestos Related Diseases, Perth, WA, Australia
| | - Alison M McDonnell
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia; National Centre for Asbestos Related Diseases, Perth, WA, Australia
| | - Richard A Lake
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia; National Centre for Asbestos Related Diseases, Perth, WA, Australia
| | - Anna K Nowak
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia; National Centre for Asbestos Related Diseases, Perth, WA, Australia
| |
Collapse
|
329
|
Miyashita M, Sasano H, Tamaki K, Hirakawa H, Takahashi Y, Nakagawa S, Watanabe G, Tada H, Suzuki A, Ohuchi N, Ishida T. Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: a retrospective multicenter study. Breast Cancer Res 2015; 17:124. [PMID: 26341640 PMCID: PMC4560879 DOI: 10.1186/s13058-015-0632-x] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
Introduction The status of tumor-infiltrating lymphocytes (TILs) has been recently proposed to predict clinical outcome of patients with breast cancer. We therefore studied the prognostic significance of CD8+ TILs and FOXP3+ TILs in residual tumors after neoadjuvant chemotherapy (NAC) and the alterations in these parameters before and after NAC in patients with triple-negative breast cancer (TNBC). Methods One hundred thirty-one TNBC patients who received NAC at three institutions were examined. CD8+ TIL and FOXP3+ TIL in residual tumors and biopsy specimens were evaluated by double-staining immunohistochemistry. The CD8+ TIL and FOXP3+ TIL status of the residual tumors was assessed, and the rates of their changes before and after NAC were calculated. Results TNBC patients with high CD8+ TIL levels or a high CD8/FOXP3 ratio in residual tumors had significantly better recurrence-free survival (RFS) and breast cancer-specific survival (BCSS) than patients with low values of these parameters. In multivariate analyses, CD8+ TIL exhibited strong prognostic significance for RFS, with a hazard ratio (HR) of 3.09 (95 % confidence interval (CI) 1.537–6.614, P=0.0013). The CD8/FOXP3 ratio was also significantly correlated with RFS (HR=2.07, 95 % CI 1.029–4.436, P=0.0412). TNBC with larger residual tumor size and positive lymph node status, which are known prognostic factors, was independently associated with worse RFS (P=0.0064 and P=0.0015, respectively). High CD8+ TIL levels were a markedly powerful indicator of improved BCSS, with an HR of 3.59 (95 % CI 1.499–9.581, P=0.0036). Nodal status was also associated with BCSS (P=0.0024). TNBC with a high rate of CD8+ TIL changes was associated with significantly better RFS compared with the low group (P=0.011). Higher rates of changes in the CD8/FOXP3 ratio were significantly correlated with both better RFS and BCSS compared with lower rates (P=0.011 and P=0.023, respectively). Conclusions This is the first study to demonstrate that high CD8+ TIL and a high CD8/FOXP3 ratio in residual tumors and increment of these parameters following NAC and accurately predict improved prognosis in TNBC patients with non-pathological complete response following NAC. These parameters could serve as a surrogate one for adjuvant treatment in patients with residual disease in the neoadjuvant setting. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0632-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minoru Miyashita
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan. .,Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Kentaro Tamaki
- Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan. .,Department of Breast Surgery, Nahanishi Clinic, 2-1-9 Akamine, Naha, 901-0154, Japan.
| | - Hisashi Hirakawa
- Department of Breast Surgery, Tohoku Kosai Hospital, 2-3-11 Kokubuncho, Aoba-ku, Sendai, 980-0803, Japan.
| | - Yayoi Takahashi
- Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Saki Nakagawa
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan. .,Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Gou Watanabe
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Hiroshi Tada
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Akihiko Suzuki
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Noriaki Ohuchi
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Takanori Ishida
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| |
Collapse
|
330
|
Ibana JA, Cutay SJ, Romero M, Schust DJ. Parallel Expression of Enzyme Inhibitors of CD8T Cell Activity in Tumor Microenvironments and Secretory Endometrium. Reprod Sci 2015; 23:289-301. [PMID: 26335176 DOI: 10.1177/1933719115602762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The divergent requirement for tolerance to support conception and protective response against sexually transmitted infections defines the unique immunological dynamics in the female reproductive tract (FRT). In part, these requirements are achieved by the cyclic modulation of cytolytic CD8T cell function in the FRT that underlie the respective immunosuppressive and immunocompetent milieus during the secretory and proliferative phases of the menstrual cycle. The CD8T cell function can be dampened by exposure to indoleamine 2,3-dioxygenase and/or arginase enzymes. Indeed, these 2 enzymes are known as primary inducers of immune suppression in tumor microenvironments. This review discusses the intriguing parallel expression of these 2 enzymes in tumor microenvironments and in the secretory endometrium. We surmise that investigating the underlying natural mechanisms that suppress and restore the immunocompetence of CD8T cells in the FRT each month may provide valuable insights into ways to artificially recapitulate these mechanisms and inhibit immune suppression in tumor microenvironments.
Collapse
Affiliation(s)
- Joyce A Ibana
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, Philippines Natural Sciences Research Institute, University of the Philippines, Diliman, Quezon City, Philippines
| | - Sandra Jelyn Cutay
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, Philippines
| | - Maevel Romero
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, Philippines Natural Sciences Research Institute, University of the Philippines, Diliman, Quezon City, Philippines
| | - Danny Joseph Schust
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
331
|
Lozano T, Villanueva L, Durántez M, Gorraiz M, Ruiz M, Belsúe V, Riezu-Boj JI, Hervás-Stubbs S, Oyarzábal J, Bandukwala H, Lourenço AR, Coffer PJ, Sarobe P, Prieto J, Casares N, Lasarte JJ. Inhibition of FOXP3/NFAT Interaction Enhances T Cell Function after TCR Stimulation. THE JOURNAL OF IMMUNOLOGY 2015; 195:3180-9. [DOI: 10.4049/jimmunol.1402997] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/27/2015] [Indexed: 01/13/2023]
|
332
|
Treg Cell Differentiation: From Thymus to Peripheral Tissue. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:175-205. [PMID: 26615097 DOI: 10.1016/bs.pmbts.2015.07.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Regulatory T cells (Tregs) are crucial mediators of self-tolerance in the periphery. They differentiate in the thymus, where interactions with thymus-resident antigen-presenting cells, an instructive cytokine milieu, and stimulation of the T cell receptor lead to the selection into the Treg lineage and the induction of Foxp3 gene expression. Once mature, Treg cells leave the thymus and migrate into either the secondary lymphoid tissues, e.g., lymph nodes and spleen, or peripheral nonlymphoid tissues. There is growing evidence that Treg cells go beyond the classical modulation of immune responses and also play important functional roles in nonlymphoid peripheral tissues. In this review, we summarize recent findings about the thymic Treg lineage differentiation as well as the further specialization of Treg cells in the secondary lymphoid and in the peripheral nonlymphoid organs.
Collapse
|
333
|
Li L, Goedegebuure SP, Fleming TP, Gillanders WE. Developing a clinical development paradigm for translation of a mammaglobin-A DNA vaccine. Immunotherapy 2015; 7:709-11. [PMID: 26250406 DOI: 10.2217/imt.15.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Lijin Li
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA.,The Alvin J Siteman Cancer Center at Barnes-Jewish Hospital & Washington University School of Medicine, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA.,The Alvin J Siteman Cancer Center at Barnes-Jewish Hospital & Washington University School of Medicine, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA.,The Alvin J Siteman Cancer Center at Barnes-Jewish Hospital & Washington University School of Medicine, MO, USA
| |
Collapse
|
334
|
Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, Diehn M, West RB, Plevritis SK, Alizadeh AA. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med 2015; 21:938-945. [PMID: 26193342 PMCID: PMC4852857 DOI: 10.1038/nm.3909] [Citation(s) in RCA: 2311] [Impact Index Per Article: 231.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/19/2015] [Indexed: 12/12/2022]
Abstract
Molecular profiles of tumors and tumor-associated cells hold great promise as biomarkers of clinical outcomes. However, existing data sets are fragmented and difficult to analyze systematically. Here we present a pan-cancer resource and meta-analysis of expression signatures from ∼18,000 human tumors with overall survival outcomes across 39 malignancies. By using this resource, we identified a forkhead box MI (FOXM1) regulatory network as a major predictor of adverse outcomes, and we found that expression of favorably prognostic genes, including KLRB1 (encoding CD161), largely reflect tumor-associated leukocytes. By applying CIBERSORT, a computational approach for inferring leukocyte representation in bulk tumor transcriptomes, we identified complex associations between 22 distinct leukocyte subsets and cancer survival. For example, tumor-associated neutrophil and plasma cell signatures emerged as significant but opposite predictors of survival for diverse solid tumors, including breast and lung adenocarcinomas. This resource and associated analytical tools (http://precog.stanford.edu) may help delineate prognostic genes and leukocyte subsets within and across cancers, shed light on the impact of tumor heterogeneity on cancer outcomes, and facilitate the discovery of biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Andrew J Gentles
- Center for Cancer Systems Biology (CCSB), Stanford University, Stanford, California, USA
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Chih Long Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Scott V Bratman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Weiguo Feng
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Dongkyoon Kim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Viswam S Nair
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Yue Xu
- Department of Cardiothoracic Surgery, Division of Thoracic Surgery, Stanford University, Stanford, California, USA
| | - Amanda Khuong
- Department of Cardiothoracic Surgery, Division of Thoracic Surgery, Stanford University, Stanford, California, USA
| | - Chuong D Hoang
- Department of Cardiothoracic Surgery, Division of Thoracic Surgery, Stanford University, Stanford, California, USA
| | - Maximilian Diehn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Robert B West
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Sylvia K Plevritis
- Center for Cancer Systems Biology (CCSB), Stanford University, Stanford, California, USA
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Ash A Alizadeh
- Center for Cancer Systems Biology (CCSB), Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
- Department of Medicine, Division of Hematology, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
335
|
Tiriveedhi V, Tucker N, Herndon J, Li L, Sturmoski M, Ellis M, Ma C, Naughton M, Lockhart AC, Gao F, Fleming T, Goedegebuure P, Mohanakumar T, Gillanders WE. Safety and preliminary evidence of biologic efficacy of a mammaglobin-a DNA vaccine in patients with stable metastatic breast cancer. Clin Cancer Res 2015; 20:5964-75. [PMID: 25451106 DOI: 10.1158/1078-0432.ccr-14-0059] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Mammaglobin-A (MAM-A) is overexpressed in 40% to 80% of primary breast cancers. We initiated a phase I clinical trial of a MAM-A DNA vaccine to evaluate its safety and biologic efficacy. EXPERIMENTAL DESIGN Patients with breast cancer with stable metastatic disease were eligible for enrollment. Safety was monitored with clinical and laboratory assessments. The CD8 T-cell response was measured by ELISPOT, flow cytometry, and cytotoxicity assays. Progression-free survival (PFS) was described using the Kaplan-Meier product limit estimator. RESULTS Fourteen subjects have been treated with the MAM-A DNA vaccine and no significant adverse events have been observed. Eight of 14 subjects were HLA-A2(+), and the CD8 T-cell response to vaccination was studied in detail. Flow cytometry demonstrated a significant increase in the frequency of MAM-A-specific CD8 T cells after vaccination (0.9% ± 0.5% vs. 3.8% ± 1.2%; P < 0.001), and ELISPOT analysis demonstrated an increase in the number of MAM-A-specific IFNγ-secreting T cells (41 ± 32 vs. 215 ± 67 spm; P < 0.001). Although this study was not powered to evaluate progression-free survival (PFS), preliminary evidence suggests that subjects treated with the MAM-A DNA vaccine had improved PFS compared with subjects who met all eligibility criteria, were enrolled in the trial, but were not vaccinated because of HLA phenotype. CONCLUSION The MAM-A DNA vaccine is safe, capable of eliciting MAM-A-specific CD8 T-cell responses, and preliminary evidence suggests improved PFS. Additional studies are required to define the potential of the MAM-A DNA vaccine for breast cancer prevention and/or therapy.
Collapse
Affiliation(s)
- Venkataswarup Tiriveedhi
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. Department of Biological Sciences, Tennessee State University, Nashville, Tennessee
| | - Natalia Tucker
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - John Herndon
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Sturmoski
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew Ellis
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - Cynthia Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - Michael Naughton
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - A Craig Lockhart
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - Feng Gao
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri. Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy Fleming
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| | - Thalachallour Mohanakumar
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri. Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
336
|
Wolf D, Sopper S, Pircher A, Gastl G, Wolf AM. Treg(s) in Cancer: Friends or Foe? J Cell Physiol 2015; 230:2598-605. [DOI: 10.1002/jcp.25016] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Dominik Wolf
- Medical Clinic 3; Oncology; Hematology and Rheumatology; University Hospital Bonn (UKB); Bonn Germany
- Department of Hematology and Oncology; Internal Medicine 5; Medical University Innsbruck; Innsbruck Austria
| | - Sieghart Sopper
- Department of Hematology and Oncology; Internal Medicine 5; Medical University Innsbruck; Innsbruck Austria
- Tyrolean Cancer Research Institute (TKFI); Medical University Innsbruck; Innsbruck Austria
| | - Andreas Pircher
- Department of Hematology and Oncology; Internal Medicine 5; Medical University Innsbruck; Innsbruck Austria
| | - Guenther Gastl
- Department of Hematology and Oncology; Internal Medicine 5; Medical University Innsbruck; Innsbruck Austria
| | - Anna Maria Wolf
- Medical Clinic 3; Oncology; Hematology and Rheumatology; University Hospital Bonn (UKB); Bonn Germany
- Department of Hematology and Oncology; Internal Medicine 5; Medical University Innsbruck; Innsbruck Austria
| |
Collapse
|
337
|
Halvorsen EC, Mahmoud SM, Bennewith KL. Emerging roles of regulatory T cells in tumour progression and metastasis. Cancer Metastasis Rev 2015; 33:1025-41. [PMID: 25359584 DOI: 10.1007/s10555-014-9529-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The metastasis of cancer is a complex and life-threatening process that is only partially understood. Immune suppressive cells are recognized as important contributors to tumour progression and may also promote the development and growth of tumour metastases. Specifically, regulatory T cells (Tregs) have been found to promote primary tumour progression, and emerging pre-clinical data suggests that Tregs may promote metastasis and metastatic tumour growth. While the precise role that Tregs play in metastatic progression is understudied, recent findings have indicated that by suppressing innate and adaptive anti-tumour immunity, Tregs may shield tumour cells from immune detection, and thereby allow tumour cells to survive, proliferate and acquire characteristics that facilitate dissemination. This review will highlight our current understanding of Tregs in metastasis, including an overview of pre-clinical findings and discussion of clinical data regarding Tregs and therapeutic outcome. Evolving strategies to directly ablate Tregs or to inhibit their function will also be discussed. Improving our understanding of how Tregs may influence tumour metastasis may lead to novel treatments for metastatic cancer.
Collapse
Affiliation(s)
- Elizabeth C Halvorsen
- Department of Integrative Oncology, British Columbia Cancer Agency, 9-202, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | | | | |
Collapse
|
338
|
Lipson EJ, Sharfman WH, Chen S, McMiller TL, Pritchard TS, Salas JT, Sartorius-Mergenthaler S, Freed I, Ravi S, Wang H, Luber B, Sproul JD, Taube JM, Pardoll DM, Topalian SL. Safety and immunologic correlates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J Transl Med 2015; 13:214. [PMID: 26143264 PMCID: PMC4491237 DOI: 10.1186/s12967-015-0572-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/11/2015] [Indexed: 02/04/2023] Open
Abstract
Background Limited adjuvant treatment options exist for patients with high-risk surgically resected melanoma. This first-in-human study investigated the safety, tolerability and immunologic correlates of Melanoma GVAX, a lethally irradiated granulocyte–macrophage colony stimulating factor (GM-CSF)-secreting allogeneic whole-cell melanoma vaccine, administered in the adjuvant setting. Methods Patients with stage IIB-IV melanoma were enrolled following complete surgical resection. Melanoma GVAX was administered intradermally once every 28 days for four cycles, at 5E7 cells/cycle (n = 3), 2E8 cells/cycle (n = 9), or 2E8 cells/cycle preceded by cyclophosphamide 200 mg/m2 to deplete T regulatory cells (Tregs; n = 8). Blood was collected before each vaccination and at 4 and 6 months after treatment initiation for immunologic studies. Vaccine injection site biopsies and additional blood samples were obtained 2 days after the 1st and 4th vaccines. Results Among 20 treated patients, 18 completed 4 vaccinations. Minimal treatment-related toxicity was observed. One patient developed vitiligo and patches of white hair during the treatment and follow-up period. Vaccine site biopsies demonstrated complex inflammatory infiltrates, including significant increases in eosinophils and PD-1+ lymphocytes from cycle 1 to cycle 4 (P < 0.05). Serum GM-CSF concentrations increased significantly in a dose-dependent manner 48 h after vaccination (P = 0.0086), accompanied by increased numbers of activated circulating monocytes (P < 0.0001) and decreased percentages of myeloid-derived suppressor cells among monocytes (CD14+ , CD11b+ , HLA-DR low or negative; P = 0.002). Cyclophosphamide did not affect numbers of circulating Tregs. No significant changes in anti-melanoma immunity were observed in peripheral T cells by interferon-gamma ELIPSOT, or immunoglobulins by serum Western blotting. Conclusion Melanoma GVAX was safe and tolerable in the adjuvant setting. Pharmacodynamic testing revealed complex vaccine site immune infiltrates and an immune-reactive profile in circulating monocytic cell subsets. These findings support the optimization of Melanoma GVAX with additional monocyte and dendritic cell activators, and the potential development of combinatorial treatment regimens with synergistic agents. Trial registration: NCT01435499 Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0572-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, 1550 Orleans Street, Room 507, Baltimore, MD, 21287, USA.
| | - William H Sharfman
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Shuming Chen
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Tracee L McMiller
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Theresa S Pritchard
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - January T Salas
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Susan Sartorius-Mergenthaler
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Irwin Freed
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Sowmya Ravi
- Department of Dermatology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Hao Wang
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Brandon Luber
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Janice Davis Sproul
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA. .,Department of Pathology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
339
|
Zhifu Y, Mingli J, Shuang C, Fan W, Zhenkun F, Wangyang C, Lin Z, Guangxiao L, Yashuang Z, Dianjun L. SNP–SNP interactions of immunity related genes involved in the CD28/B7 pathway with susceptibility to invasive ductal carcinoma of the breast. Gene 2015; 566:217-22. [DOI: 10.1016/j.gene.2015.04.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/27/2015] [Accepted: 04/17/2015] [Indexed: 12/16/2022]
|
340
|
Winkler I, Wilczynska B, Bojarska-Junak A, Gogacz M, Adamiak A, Postawski K, Darmochwal-Kolarz D, Rechberger T, Tabarkiewicz J. Regulatory T lymphocytes and transforming growth factor beta in epithelial ovarian tumors-prognostic significance. J Ovarian Res 2015; 8:39. [PMID: 26077607 PMCID: PMC4513978 DOI: 10.1186/s13048-015-0164-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 06/01/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Regulatory T lymphocytes (Treg) are characterized by the presence of CD4+ surface antigen. Today the transcription factor FOXP3 is considered to be the most specific marker of Treg cells. The aim of the study was to estimate the percentage of Treg in peripheral blood and the tissue of the epithelial ovarian tumor and blood serum TGF-beta concentrations and relationships between them. Moreover, the aim of the study was to answer the question whether the percentage of Treg lymphocytes affects the time of survival in patients with ovarian cancer. METHODS The patients were divided into four groups, depending on the histopathological examination result: I--a group without any pathology within the ovaries (C; n = 20), II--a group with benign tumors (B; n = 25), III - with borderline tumors (BR; n = 11), IV--a group with cancer of the ovary (M; n = 24). The percentage of Treg lymphocytes in peripheral blood and the tissue was assessed using the flow cytometry method. TGF-beta cytokine concentration was estimated with the ELISA immunoenzymatic test. Statistical analysis of the results was conducted using the computer program Statistica 10.0PL (StatSoft, Inc). RESULTS No significant differences were found in percentages of Treg lymphocytes in peripheral blood between individual groups of patients (p = 0.11). However, we observed marked differences in the tissue of malignant and non-malignant tumors between individual groups of patients (p = 0.003). The analysis with the post hoc test revealed significantly higher TGF-beta concentration in the group of women with malignant tumors. Moreover, no relationship was found between TGF-beta concentration and the percentage of Treg cells in peripheral blood and tumors of the ovary. No correlation was found between the percentage of Treg lymphocytes in peripheral blood (p = 0.4) and the tissue of ovarian tumors (p = 0.3) and the time of survival of patients with ovarian cancer. CONCLUSIONS The recruitment of Treg lymphocytes toward the tumor is one of the mechanisms of escape of neoplasm from the response of the immune system. The percentage of Treg lymphocytes in peripheral blood and the neoplastic tissue does not influence the time of survival of patients with ovarian cancer.
Collapse
Affiliation(s)
- Izabela Winkler
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Barbara Wilczynska
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland. .,Department of Paediatric Endocrinology and Diabetology with Endocrine-Metabolic Laboratory, Chodźki 2 Street, 20-093, Lublin, Poland.
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland.
| | - Marek Gogacz
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Aneta Adamiak
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Krzysztof Postawski
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Dorota Darmochwal-Kolarz
- Department of Obstetrics and Perinatology, Medical University of Lublin, Jaczewski Street, 20-954, Lublin, Poland. .,Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| | - Tomasz Rechberger
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| |
Collapse
|
341
|
Abstract
A number of consensuses regarding cancer immunology have recently emerged from both preclinical immunotherapy models and analysis of cancer patients. First and foremost, the natural state of endogenous tumor reactive T cells is characterized by general hyporesponsiveness or anergy. This is likely due to a number of mechanisms that tumors use to induce tolerance as they develop. While many of the newer generation vaccines can effectively transfer antigen to and activate dendritic cells, T-cell tolerance remains a major barrier that is difficult to overcome by vaccination alone. Preclinical models demonstrate that for poorly immunogenic tumors, once tolerance has been established, therapeutic vaccines alone are ineffective at curing animals with a significant established tumor burden. However, combination strategies of vaccination together with inhibitors of immunologic checkpoints and agonists for co-stimulatory pathways are proving capable of overcoming tolerance and generating significant anti-tumor responses even in cases of established metastatic cancer.
Collapse
Affiliation(s)
- Drew Pardoll
- Department of Oncology and Director Cancer Immunology Program, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
342
|
Aguilar LK, Shirley LA, Chung VM, Marsh CL, Walker J, Coyle W, Marx H, Bekaii-Saab T, Lesinski GB, Swanson B, Sanchez D, Manzanera AG, Aguilar-Cordova E, Bloomston M. Gene-mediated cytotoxic immunotherapy as adjuvant to surgery or chemoradiation for pancreatic adenocarcinoma. Cancer Immunol Immunother 2015; 64:727-36. [PMID: 25795132 PMCID: PMC11029723 DOI: 10.1007/s00262-015-1679-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/04/2015] [Indexed: 01/30/2023]
Abstract
BACKGROUND While surgical resection of pancreatic adenocarcinoma provides the only chance of cure, long-term survival remains poor. Immunotherapy may improve outcomes, especially as adjuvant to local therapies. Gene-mediated cytotoxic immunotherapy (GMCI) generates a systemic anti-tumor response through local delivery of an adenoviral vector expressing the HSV-tk gene (aglatimagene besadenovec, AdV-tk) followed by anti-herpetic prodrug. GMCI has demonstrated synergy with standard of care (SOC) in other tumor types. This is the first application in pancreatic cancer. METHODS Four dose levels (3 × 10(10) to 1 × 10(12) vector particles) were evaluated as adjuvant to surgery for resectable disease (Arm A) or to 5-FU chemoradiation for locally advanced disease (Arm B). Each patient received two cycles of AdV-tk + prodrug. RESULTS Twenty-four patients completed therapy, 12 per arm, with no dose-limiting toxicities. All Arm A patients were explored, eight were resected, one was locally advanced and three had distant metastases. CD8(+) T cell infiltration increased an average of 22-fold (range sixfold to 75-fold) compared with baseline (p = 0.0021). PD-L1 expression increased in 5/7 samples analyzed. One node-positive resected patient is alive >66 months without recurrence. Arm B RECIST response rate was 25 % with a median OS of 12 months and 1-year survival of 50 %. Patient-reported quality of life showed no evidence of deterioration. CONCLUSIONS AdV-tk can be safely combined with pancreatic cancer SOC without added toxicity. Response and survival compare favorably to expected outcomes and immune activity increased. These results support further evaluation of GMCI with more modern chemoradiation and surgery as well as PD-1/PD-L1 inhibitors in pancreatic cancer.
Collapse
Affiliation(s)
| | - Lawrence A. Shirley
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| | | | | | - Jon Walker
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| | | | - Howard Marx
- City of Hope National Medical Center, Duarte, CA 91010 USA
| | - Tanios Bekaii-Saab
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| | - Gregory B. Lesinski
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| | - Benjamin Swanson
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| | | | | | | | - Mark Bloomston
- James Cancer Hospital/Solove Research Institute, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
343
|
Yang WY, Shao Y, Lopez-Pastrana J, Mai J, Wang H, Yang XF. Pathological conditions re-shape physiological Tregs into pathological Tregs. BURNS & TRAUMA 2015; 3. [PMID: 26623425 PMCID: PMC4662545 DOI: 10.1186/s41038-015-0001-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4+FOXP3+ regulatory T cells (Tregs) are a subset of CD4 T cells that play an essential role in maintaining peripheral immune tolerance, controlling acute and chronic inflammation, allergy, autoimmune diseases, and anti-cancer immune responses. Over the past 20 years, significant progress has been made since Tregs were first characterized in 1995. Many concepts and principles regarding Tregs generation, phenotypic features, subsets (tTregs, pTregs, iTregs, and iTreg35), tissue specificity (central Tregs, effector Tregs, and tissue resident Tregs), homeostasis (highly dynamic and apoptotic), regulation of Tregs by receptors for PAMPs and DAMPs, Treg plasticity (re-differentiation to other CD4 T helper cell subsets, Th1, Th2, Tfh and Th17), and epigenetic regulation of Tregs phenotypes and functions have been innovated. In this concise review, we want to briefly analyze these eight new progresses in the study of Tregs. We have also proposed for the first time a novel concept that "physiological Tregs" have been re-shaped into "pathological Tregs" in various pathological environments. Continuing of the improvement in our understanding on this important cellular component about the immune tolerance and immune suppression, would lead to the future development of novel therapeutics approaches for acute and chronic inflammatory diseases, allergy, allogeneic transplantation-related immunity, sepsis, autoimmune diseases, and cancers.
Collapse
Affiliation(s)
- William Y Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Ying Shao
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Jahaira Lopez-Pastrana
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Jietang Mai
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Xiao-Feng Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A ; Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| |
Collapse
|
344
|
Zelig U, Barlev E, Bar O, Gross I, Flomen F, Mordechai S, Kapelushnik J, Nathan I, Kashtan H, Wasserberg N, Madhala-Givon O. Early detection of breast cancer using total biochemical analysis of peripheral blood components: a preliminary study. BMC Cancer 2015; 15:408. [PMID: 25975566 PMCID: PMC4455613 DOI: 10.1186/s12885-015-1414-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/05/2015] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Most of the blood tests aiming for breast cancer screening rely on quantification of a single or few biomarkers. The aim of this study was to evaluate the feasibility of detecting breast cancer by analyzing the total biochemical composition of plasma as well as peripheral blood mononuclear cells (PBMCs) using infrared spectroscopy. METHODS Blood was collected from 29 patients with confirmed breast cancer and 30 controls with benign or no breast tumors, undergoing screening for breast cancer. PBMCs and plasma were isolated and dried on a zinc selenide slide and measured under a Fourier transform infrared (FTIR) microscope to obtain their infrared absorption spectra. Differences in the spectra of PBMCs and plasma between the groups were analyzed as well as the specific influence of the relevant pathological characteristics of the cancer patients. RESULTS Several bands in the FTIR spectra of both blood components significantly distinguished patients with and without cancer. Employing feature extraction with quadratic discriminant analysis, a sensitivity of ~90 % and a specificity of ~80 % for breast cancer detection was achieved. These results were confirmed by Monte Carlo cross-validation. Further analysis of the cancer group revealed an influence of several clinical parameters, such as the involvement of lymph nodes, on the infrared spectra, with each blood component affected by different parameters. CONCLUSION The present preliminary study suggests that FTIR spectroscopy of PBMCs and plasma is a potentially feasible and efficient tool for the early detection of breast neoplasms. An important application of our study is the distinction between benign lesions (considered as part of the non-cancer group) and malignant tumors thus reducing false positive results at screening. Furthermore, the correlation of specific spectral changes with clinical parameters of cancer patients indicates for possible contribution to diagnosis and prognosis.
Collapse
Affiliation(s)
- Udi Zelig
- Todos Medical Ltd, 1 HaMada St, Rehovot, 76703, Israel.
| | - Eyal Barlev
- Department Surgery B, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Omri Bar
- Todos Medical Ltd, 1 HaMada St, Rehovot, 76703, Israel.
| | - Itai Gross
- Pediatric Hemato-Oncology Unit, Soroka University Medical Center and Faculty of Medicine, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Felix Flomen
- Todos Medical Ltd, 1 HaMada St, Rehovot, 76703, Israel.
| | - Shaul Mordechai
- Department of Physics, Ben Gurion University, Beer-Sheva, Israel.
| | - Joseph Kapelushnik
- Pediatric Hemato-Oncology Unit, Soroka University Medical Center and Faculty of Medicine, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Ilana Nathan
- Department Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, and Institute of Hematology, Soroka University Medical Center, Beer-Sheva, Israel.
| | - Hanoch Kashtan
- Division of General Surgery, Rabin Medical Center, Campus Beilinson, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv, Israel.
| | - Nir Wasserberg
- Department Surgery B, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Osnat Madhala-Givon
- Department Surgery B, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
345
|
Sawant DV, Hamilton K, Vignali DAA. Interleukin-35: Expanding Its Job Profile. J Interferon Cytokine Res 2015; 35:499-512. [PMID: 25919641 DOI: 10.1089/jir.2015.0015] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Counter-regulation afforded by specialized regulatory cell populations and immunosuppressive cytokines is critical for balancing immune outcome. The inhibitory potential of the established suppressive cytokines, IL-10 and TGFβ, has been well elucidated in diverse inflammatory scenarios in conjunction with their key roles in Treg development and function. Despite the early predictions for an immunomodulatory role for the Ebi3/p35 heterodimer in placental trophoblasts, IL-35 biology remained elusive until 2007 when it was established as a Treg-restricted inhibitory cytokine. Since then, Treg-derived IL-35 has been shown to exhibit its suppressive activities in a range of autoimmune diseases and cancer models. Recent studies are beginning to explore other cellular sources of IL-35, such as Bregs and CD8(+) Tregs. Despite these new cellular sources and targets, the mode of IL-35 suppression remains restricted to inhibition of proliferation and induction of an IL-35-producing induced regulatory T cell population referred to as iTr35. In this review, we explore the early beginnings, status quo, and future prospects of IL-35 biology. The unparalleled opportunity of targeting multiple immunosuppressive populations (Tregs, Bregs, CD8(+) Tregs) through IL-35 is highly exciting and offers tremendous promise from a translational standpoint, particularly for cancer immunotherapies.
Collapse
Affiliation(s)
- Deepali V Sawant
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Kristia Hamilton
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
346
|
Xuan W, Yan Y, Wan M, Wu X, Ji D, Wang L, Lin C, Chen Y, Yu Y, Zhang X. Antitumor activity of mHSP65-TTL enhanced by administration of low dose cyclophosphamide in pancreatic cancer-bearing mice. Int Immunopharmacol 2015; 27:95-103. [PMID: 25907246 DOI: 10.1016/j.intimp.2015.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer remains a lethal malignancy. Despite chemotherapy or/and radiotherapy after the surgery, the improvement on the overall survival of the patients has still been minimal. To develop novel therapeutic approaches, we tried to prepare mHSP65-TTL, a candidate vaccine prepared by mixing the recombinant mycobacterial heat shock protein 65 (mHSP65) with tumor tissue lysate (TTL) of Panc02 pancreatic cancer tissue. The mHSP65-TTL were used to immune the C57BL/6 mice implanted with the Panc02 cancer cells, in combination with or without low dose cyclophosphamide (CY). The results showed that mHSP65-TTL significantly prolonged the survival of the pancreatic cancer bearing mice and low dose CY enhanced the efficacy of the mHSP65-TTL. In addition, we detected mRNA expression of RORγt and IL-17A in spleen cells of mice received mHSP65-TTL or mHSP65-TTL plus CY, and found that mHSP65-TTL up-regulated mRNA expressions of RORγt and IL-17A, CY alone or mHSP65-TTL plus CY up-regulated mRNA expressions of RORγt. The work could provide an insight into a combinational approach for the immunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Youyou Yan
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Min Wan
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Xiuli Wu
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Degang Ji
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Liying Wang
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Chao Lin
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Yang Chen
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Yongli Yu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China.
| | - Xuewen Zhang
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China.
| |
Collapse
|
347
|
Karimi S, Chattopadhyay S, Chakraborty NG. Manipulation of regulatory T cells and antigen-specific cytotoxic T lymphocyte-based tumour immunotherapy. Immunology 2015; 144:186-96. [PMID: 25243729 DOI: 10.1111/imm.12387] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 12/16/2022] Open
Abstract
The most potent killing machinery in our immune system is the cytotoxic T lymphocyte (CTL). Since the possibility for self-destruction by these cells is high, many regulatory activities exist to prevent autoimmune destruction by these cells. A tumour (cancer) grows from the cells of the body and is tolerated by the body's immune system. Yet, it has been possible to generate tumour-associated antigen (TAA) -specific CTL that are also self-antigen specific in vivo, to achieve a degree of therapeutic efficacy. Tumour-associated antigen-specific T-cell tolerance through pathways of self-tolerance generation represents a significant challenge to successful immunotherapy. CD4(+) CD25(+) FoxP3(+) T cells, referred to as T regulatory (Treg) cells, are selected in the thymus as controllers of the anti-self repertoire. These cells are referred to as natural T regulatory (nTreg) cells. According to the new consensus (Nature Immunology 2013; 14:307-308) these cells are to be termed as (tTreg). There is another class of CD4(+) Treg cells also involved in regulatory function in the periphery, also phenotypically CD4(+) CD25(±) , classified as induced Treg (iTreg) cells. These cells are to be termed as peripherally induced Treg (pTreg) cells. In vitro-induced Treg cells with suppressor function should be termed as iTreg. These different Treg cells differ in their requirements for activation and in their mode of action. The current challenges are to determine the degree of specificity of these Treg cells in recognizing the same TAA as the CTL population and to circumvent their regulatory constraints so as to achieve robust CTL responses against cancer.
Collapse
Affiliation(s)
- Shirin Karimi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
348
|
Watanabe R, Yasuno T, Hisano S, Sasatomi Y, Nakashima H. Distinct cytokine mRNA expression pattern in immunoglobulin G4-related kidney disease associated with renal cell carcinoma. Clin Kidney J 2015; 7:269-74. [PMID: 25852888 PMCID: PMC4377746 DOI: 10.1093/ckj/sfu024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/21/2014] [Indexed: 12/24/2022] Open
Abstract
We treated a 61-year-old man with immunoglobulin (Ig)G4-related kidney disease (IgG4-RKD). He had a history of allergic diseases and an allergic reaction and had received a diagnosis of autoimmune pancreatitis (AIP). He had also received a diagnosis of renal cell carcinoma (RCC) and had undergone segmental resection of the left kidney at 59 years of age. His serum amylase level and number of peripheral eosinophils increased after RCC development. We hypothesized that the RCC may have induced AIP and IgG4-RKD and we therefore examined the excised RCC tissue; typical findings of IgG4-RKD associated with RCC were recognized. We next evaluated the mRNA expression of cytokines in the excised tissues of this case and ten other ordinary RCC cases. In all cases, notable levels of IL-10 mRNA and high levels of TGF-β mRNA were seen. Although prominent differences were not observed in the mRNA expression of Th1, Th17 and Treg cytokines in all cases, the present case alone showed increased production of the Th2 cytokines IL-4 and IL-5, which were not detected in ordinary RCC cases. Although the mechanism underlying IgG4-RKD development has not yet been determined, Th2 and Treg cells are thought to play a prominent role in the pathogenesis. It is therefore likely that in this case, the association of these two diseases was not coincidental, and a distinct immune response against RCC may trigger IgG4-RKD development.
Collapse
Affiliation(s)
- Renya Watanabe
- Division of Nephrology and Rheumatology, Department of Internal Medicine , Fukuoka University , Fukuoka , Japan
| | - Tetsuhiko Yasuno
- Division of Nephrology and Rheumatology, Department of Internal Medicine , Fukuoka University , Fukuoka , Japan
| | - Satoshi Hisano
- Department of Pathology, Faculty of Medicine , Fukuoka University , Fukuoka , Japan
| | - Yoshie Sasatomi
- Division of Nephrology and Rheumatology, Department of Internal Medicine , Fukuoka University , Fukuoka , Japan
| | - Hitoshi Nakashima
- Division of Nephrology and Rheumatology, Department of Internal Medicine , Fukuoka University , Fukuoka , Japan
| |
Collapse
|
349
|
Datta J, Rosemblit C, Berk E, Showalter L, Namjoshi P, Mick R, Lee KP, Brod AM, Yang RL, Kelz RR, Fitzpatrick E, Hoyt C, Feldman MD, Zhang PJ, Xu S, Koski GK, Czerniecki BJ. Progressive loss of anti-HER2 CD4 + T-helper type 1 response in breast tumorigenesis and the potential for immune restoration. Oncoimmunology 2015; 4:e1022301. [PMID: 26451293 DOI: 10.1080/2162402x.2015.1022301] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/15/2015] [Accepted: 02/17/2015] [Indexed: 01/01/2023] Open
Abstract
Genomic profiling has identified several molecular oncodrivers in breast tumorigenesis. A thorough understanding of endogenous immune responses to these oncodrivers may provide insights into immune interventions for breast cancer (BC). We investigated systemic anti-HER2/neu CD4+ T-helper type-1 (Th1) responses in HER2-driven breast tumorigenesis. A highly significant stepwise Th1 response loss extending from healthy donors (HD), through HER2pos-DCIS, and ultimately to early stage HER2pos-invasive BC patients was detected by IFNγ ELISPOT. The anti-HER2 Th1 deficit was not attributable to host-level T-cell anergy, loss of immune competence, or increase in immunosuppressive phenotypes (Treg/MDSCs), but rather associated with a functional shift in IFNγ:IL-10-producing phenotypes. HER2high, but not HER2low, BC cells expressing IFNγ/TNF-α receptors were susceptible to Th1 cytokine-mediated apoptosis in vitro, which could be significantly rescued by neutralizing IFNγ and TNF-α, suggesting that abrogation of HER2-specific Th1 may reflect a mechanism of immune evasion in HER2-driven tumorigenesis. While largely unaffected by cytotoxic or HER2-targeted (trastuzumab) therapies, depressed Th1 responses in HER2pos-BC patients were significantly restored following HER2-pulsed dendritic cell (DC) vaccinations, suggesting that this Th1 defect is not "fixed" and can be corrected by immunologic interventions. Importantly, preserved anti-HER2 Th1 responses were associated with pathologic complete response to neoadjuvant trastuzumab/chemotherapy, while depressed responses were observed in patients incurring locoregional/systemic recurrence following trastuzumab/chemotherapy. Monitoring anti-HER2 Th1 reactivity following HER2-directed therapies may identify vulnerable subgroups at risk of clinicopathologic failure. In such patients, combinations of existing HER2-targeted therapies with strategies to boost anti-HER2 CD4+ Th1 immunity may decrease the risk of recurrence and thus warrant further investigation.
Collapse
Affiliation(s)
- Jashodeep Datta
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Cinthia Rosemblit
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Erik Berk
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Lori Showalter
- Department of Biological Sciences; Kent State University ; Kent, OH USA
| | - Prachi Namjoshi
- Department of Biological Sciences; Kent State University ; Kent, OH USA
| | - Rosemarie Mick
- Department of Epidemiology and Biostatistics; University of Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Kathreen P Lee
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Andrew M Brod
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Rachel L Yang
- Department of Surgery; Stanford University Hospital and Clinics ; Palo Alto, CA USA
| | - Rachel R Kelz
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Elizabeth Fitzpatrick
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Clifford Hoyt
- Life Sciences & Technology; PerkinElmer Inc. ; Hopkinton, MA USA
| | - Michael D Feldman
- Department of Pathology; University of Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Paul J Zhang
- Department of Pathology; University of Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Shuwen Xu
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA
| | - Gary K Koski
- Department of Biological Sciences; Kent State University ; Kent, OH USA
| | - Brian J Czerniecki
- Department of Surgery; University Pennsylvania Perelman School of Medicine ; Philadelphia, PA USA ; Rena Rowen Breast Center; Hospital of the University of Pennsylvania ; Philadelphia, PA USA
| |
Collapse
|
350
|
Wu J, Cui H, Zhu Z, Wang L, Li H, Wang D. Effect of HIF1α on Foxp3 expression in CD4+ CD25- T lymphocytes. Microbiol Immunol 2015; 58:409-15. [PMID: 24931519 DOI: 10.1111/1348-0421.12168] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/16/2014] [Accepted: 05/27/2014] [Indexed: 02/02/2023]
Abstract
The aim of the present study was to investigate the effect of HIF1α on Foxp3 expression in CD4(+) CD25(-) T lymphocytes. CD4(+) CD25(-) T lymphocytes were sorted from PBMC using a CD4(+) CD25(+) regulatory T cell isolation kit. Lentivirus containing lentiviral vector that overexpressed HIF1α (HIF-lenti) and those containing empty expression vector (control-lenti) were produced. Meanwhile, lentivirus that contained lentiviral vector that suppressed HIF1α expression (siHIF-lenti) and those containing control vector (sicontrol-lenti) were also generated. The sorted CD4(+) CD25(-) T lymphocytes were infected with HIF-lenti, control-lenti, siHIF-lenti, and sicontrol-lenti, respectively. Approximately 72 hr after transduction, real-time PCR and Western blot were carried out to analyze the RNA and protein expression level of HIF1α and Foxp3. CD4(+) CD25(-) T lymphocytes cultured under 21% O2 , 5% CO2 (normoxia) and 1% O2 , 5% CO2 (hypoxia) were used as control. Our results showed that overexpression of HIF1α increased both mRNA and protein expression of Foxp3 and, meanwhile, suppression of HIF1α expression by RNAi could reverse high Foxp3 expression in CD4(+) CD25(-) T lymphocytes caused by hypoxic culture. These results suggested that hypoxia could stimulate Foxp3 expression by increasing HIF1α expression in CD4(+) T lymphocytes which may promote CD4(+) T lymphocytes to convert to Treg.
Collapse
Affiliation(s)
- Jianguo Wu
- The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, 300211
| | | | | | | | | | | |
Collapse
|