1
|
Liao JB, Jejurikar NS, Hitchcock-Bernhardt KM, Gwin WR, Reichow JL, Dang Y, Childs JS, Coveler AL, Swensen RE, Goff BA, Disis ML, Salazar LG. Intraperitoneal immunotherapy with denileukin diftitox (ONTAK) in recurrent refractory ovarian cancer. Gynecol Oncol 2024; 191:74-79. [PMID: 39362046 DOI: 10.1016/j.ygyno.2024.09.019] [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: 08/09/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Denileukin diftitox (ONTAK) is a diphtheria/IL-2R fusion protein able to deplete regulatory T cells in peripheral blood. Regulatory T cells in the local immune microenvironment have been shown to be associated with poor prognosis in ovarian cancer. This study examined whether denileukin diftitox (ONTAK) could be safely administered intraperitoneal in patients with advanced refractory ovarian cancer and assessed its effects on regulatory T cells and tumor associated cytokines in ascites and peripheral blood. PATIENTS AND METHODS A phase I dose escalation study of intraperitoneal denileukin diftitox (ONTAK) enrolled 10 patients with advanced, refractory ovarian carcinoma at 3 doses (5 μg/kg, 15 μg/kg, and 25 μg/kg). Serial CA-125 measurements assessed clinical response. Regulatory T cells were quantified using RT-PCR and cytokine levels measured by Luminex. RESULTS The maximum tolerated dose was 15 μg/kg with a dose limiting toxicity observed in 1 out of 6 patients in the expansion group. The majority of adverse events were transient grades 1-2. One patient treated at the 25 μg/kg dose experienced cytokine storm with prolonged hospitalization. 3 patients had decreases in CA-125 after treatment but none met criteria for partial response. Treatment with denileukin diftitox (ONTAK) decreased regulatory T cells in peripheral blood and ascites. Treated patients did not show any significant changes in IL-8, TGF-β, sIL2Ra in ascites or peripheral blood. CONCLUSIONS Denileukin diftitox (ONTAK) can be safely administered intraperitoneally to recurrent refractory ovarian cancer patients. Regulatory T cells were reduced in ascites and peripheral blood, but there were no significant changes in cytokine levels. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov # NCT00357448.
Collapse
Affiliation(s)
- John B Liao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America.
| | - Nikita S Jejurikar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Katie M Hitchcock-Bernhardt
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - William R Gwin
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Jessica L Reichow
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Yushe Dang
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Jennifer S Childs
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Andrew L Coveler
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Ron E Swensen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America; Valley Medical Center, 400 South 43(rd) Street, Renton, WA 98055, United States of America
| | - Barbara A Goff
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America
| | - Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Lupe G Salazar
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| |
Collapse
|
2
|
Zare H, Bakherad H, Nasr Esfahani A, Norouzi M, Aghamollaei H, Mousavi Gargari SL, Mahmoodi F, Aliomrani M, Ebrahimizadeh W. Introduction of a new recombinant vaccine based on GRP78 for breast cancer immunotherapy and evaluation in a mouse model. BIOIMPACTS : BI 2023; 14:27829. [PMID: 38505675 PMCID: PMC10945302 DOI: 10.34172/bi.2023.27829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/04/2023] [Accepted: 07/29/2023] [Indexed: 03/21/2024]
Abstract
Introduction Breast cancer is one of the most prevalent malignancies in women. Several treatment options are available today, including surgery, chemotherapy, and radiotherapy. Immunotherapy, as a highly specific therapy, involves adaptive immune responses and immunological memory. In our present research, we used the recombinant C-terminal domain of the GRP78 (glucose- regulated protein 78) protein to induce an immune response and investigate its therapeutic impact in the 4T1 breast cancer model. Methods BALB/c mice were immunized with the cGRP78 protein. The humoral immune response was assessed by ELISA. Then, BALB/c mice were injected subcutaneously with 1×106 4T1 tumor cells. Subsequently, tumor size and survival rate measurements, MTT, and cytokine assays were performed. Results The animals receiving the cGRP78 vaccine showed significantly more favorable survival and slower tumor growth rates compared with unvaccinated tumor-bearing mice as the negative control mice. Circulating levels of tumoricidal cytokines such as IFNγ were higher, whereas tolerogenic cytokines such as IL-2, 6, and 10 either did not increase or had a decreasing trend in mice receiving cGRP78. Conclusion cGRP78 vaccines generated potent immunotherapeutic effects in a breast cancer mouse model. This novel strategy of targeting the GRP78 protein can promote the development of cancer vaccines and immunotherapies for breast cancer malignancies.
Collapse
Affiliation(s)
- Hamed Zare
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arman Nasr Esfahani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohamad Norouzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Mahmoodi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Mahdi Aliomrani
- Department of Pharmacology and Toxicology, Isfahan Pharmaceutical Science Research Center, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Walead Ebrahimizadeh
- Department of Surgery, Division of Urology, McGill University, and the Research Institute of the McGill University Health Centre (RI MUHC), Montreal, Quebec, Canada
| |
Collapse
|
3
|
Shan F, Somasundaram A, Bruno TC, Workman CJ, Vignali DAA. Therapeutic targeting of regulatory T cells in cancer. Trends Cancer 2022; 8:944-961. [PMID: 35853825 PMCID: PMC9588644 DOI: 10.1016/j.trecan.2022.06.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/24/2022]
Abstract
The success of immunotherapy in oncology underscores the vital role of the immune system in cancer development. Regulatory T cells (Tregs) maintain a fine balance between autoimmunity and immune suppression. They have multiple roles in the tumor microenvironment (TME) but act particularly in suppressing T cell activation. This review focuses on the detrimental and sometimes beneficial roles of Tregs in tumors, our current understanding of recruitment and stabilization of Tregs within the TME, and current Treg-targeted therapeutics. Research identifying subpopulations of Tregs and their respective functions and interactions within the complex networks of the TME will be crucial to develop the next generation of immunotherapies. Through these advances, Treg-targeted immunotherapy could have important implications for the future of oncology.
Collapse
Affiliation(s)
- Feng Shan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ashwin Somasundaram
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA.
| |
Collapse
|
4
|
Malla RR, Vasudevaraju P, Vempati RK, Rakshmitha M, Merchant N, Nagaraju GP. Regulatory T cells: Their role in triple-negative breast cancer progression and metastasis. Cancer 2022; 128:1171-1183. [PMID: 34990009 DOI: 10.1002/cncr.34084] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and immunogenic subtype of breast cancer. This tumorigenicity is independent of hormonal or HER2 pathways because of a lack of respective receptor expression. TNBC is extremely prone to drug resistance and early recurrence because of T-regulatory cell (Treg) infiltration into the tumor microenvironment (TME) in addition to other mechanisms like genomic instability. Tumor-infiltrating Tregs interact with both tumor and stromal cells as well as extracellular matrix components in the TME and induce an immune-suppressive phenotype. Hence, treatment of TNBC with conventional therapies remains challenging. Understanding the protective mechanism of Tregs in shielding TNBC from antitumor immune responses in the TME will pave the way for developing novel, immune-based therapeutics. The current review focuses on the role of tumor-infiltrating Tregs in tumor progression and metabolic reprogramming of the TME. The authors have extended their focus to oncotargeting Treg-mediated immune suppression in breast cancer. Because of its potential role in the TME, modulating Treg activity may provide a novel strategic intervention to combat TNBC. Both under laboratory conditions and in clinical trials, currently available anticancer drugs and natural therapeutics as potential agents for targeting Tregs are explored.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India.,Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Padmaraju Vasudevaraju
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Rahul Kumar Vempati
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Marni Rakshmitha
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Neha Merchant
- Department of Bioscience and Biotechnology, Banasthali University, Jaipur, India
| | | |
Collapse
|
5
|
Interleukin 2-Based Fusion Proteins for the Treatment of Cancer. J Immunol Res 2021; 2021:7855808. [PMID: 34790830 PMCID: PMC8592747 DOI: 10.1155/2021/7855808] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Interleukin 2 (IL-2) plays a fundamental role in both immune activation and tolerance and has revolutionized the field of cancer immunotherapy since its discovery. The ability of IL-2 to mediate tumor regression in preclinical and clinical settings led to FDA approval for its use in the treatment of metastatic renal cell carcinoma and metastatic melanoma in the 1990s. Although modest success is observed in the clinic, cancer patients receiving IL-2 therapy experience a wide array of side effects ranging from flu-like symptoms to life-threatening conditions such as vascular leak syndrome. Over the past three decades, efforts have focused on circumventing IL-2-related toxicities by engineering methods to localize IL-2 to the tumor or secondary lymphoid tissue, preferentially activate CD8+ T cells and NK cells, and alter pharmacokinetic properties to increase bioavailability. This review summarizes the various IL-2-based strategies that have emerged, with a focus on chimeric fusion methods.
Collapse
|
6
|
Amens JN, Bahçecioglu G, Zorlutuna P. Immune System Effects on Breast Cancer. Cell Mol Bioeng 2021; 14:279-292. [PMID: 34295441 PMCID: PMC8280260 DOI: 10.1007/s12195-021-00679-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is one of the most common cancers in women, with the ability to metastasize to secondary organs, which is the main cause of cancer-related deaths. Understanding how breast tumors progress is essential for developing better treatment strategies against breast cancer. Until recently, it has been considered that breast cancer elicits a small immune response. However, it is now clear that breast tumor progression is either prevented by the action of antitumor immunity or exacerbated by proinflammatory cytokines released mainly by the immune cells. In this comprehensive review we first explain antitumor immunity, then continue with how the tumor suppresses and evades the immune response, and next, outline the role of inflammation in breast tumor initiation and progression. We finally review the current immunotherapeutic and immunoengineering strategies against breast cancer as a promising emerging approach for the discovery and design of immune system-based strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Jensen N. Amens
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Gökhan Bahçecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
7
|
Thibodeaux SR, Barnett BB, Pandeswara S, Wall SR, Hurez V, Dao V, Sun L, Daniel BJ, Brumlik MJ, Drerup J, Padrón Á, Whiteside T, Kryczek I, Zou W, Curiel TJ. IFNα Augments Clinical Efficacy of Regulatory T-cell Depletion with Denileukin Diftitox in Ovarian Cancer. Clin Cancer Res 2021; 27:3661-3673. [PMID: 33771857 DOI: 10.1158/1078-0432.ccr-20-4594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/14/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunotherapy treats some cancers, but not ovarian cancer. Regulatory T cells (Tregs) impede anti-ovarian cancer immunity but effective human Treg-directed treatments are lacking. We tested Treg depletion with denileukin diftitox (DD) ± IFNα as ovarian cancer immunotherapy. PATIENTS AND METHODS Mice with syngeneic ID8 ovarian cancer challenge were treated with DD, IFNα, or both. The phase 0/I trial tested one dose-escalated DD infusion for functional Treg reduction, safety, and tolerability. The phase II trial added IFNα2a to DD if DD alone failed clinically. RESULTS DD depleted Tregs, and improved antitumor immunity and survival in mice. IFNα significantly improved antitumor immunity and survival with DD. IFNα did not alter Treg numbers or function but boosted tumor-specific immunity and reduced tumor Treg function with DD by inducing dendritic cell IL6. DD alone was well tolerated, depleted functional blood Tregs and improved immunity in patients with various malignancies in phase 0/I. A patient with ovarian cancer in phase 0/I experienced partial clinical response prompting a phase II ovarian cancer trial, but DD alone failed phase II. Another phase II trial added pegylated IFNα2a to failed DD, producing immunologic and clinical benefit in two of two patients before a DD shortage halt. DD alone was well tolerated. Adding IFNα increased toxicities but was tolerable, and reduced human Treg numbers in blood, and function through dendritic cell-induced IL6 in vitro. CONCLUSIONS Treg depletion is clinically useful but unlikely alone to cure ovarian cancer. Rational treatment agent combinations can salvage clinical failure of Treg depletion alone, even when neither single agent provides meaningful clinical benefit.
Collapse
Affiliation(s)
- Suzanne R Thibodeaux
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas.,Department of Medicine, University of Texas Health San Antonio, Texas
| | - Brian B Barnett
- Tulane Medical School, Department of Medicine, New Orleans, Louisiana
| | | | - Shawna R Wall
- Department of Medicine, University of Texas Health San Antonio, Texas
| | - Vincent Hurez
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas.,Department of Medicine, University of Texas Health San Antonio, Texas
| | - Vinh Dao
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas
| | - Lishi Sun
- Department of Medicine, University of Texas Health San Antonio, Texas
| | - Benjamin J Daniel
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas.,Department of Medicine, University of Texas Health San Antonio, Texas
| | - Michael J Brumlik
- Department of Medicine, University of Texas Health San Antonio, Texas
| | - Justin Drerup
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas
| | - Álvaro Padrón
- Department of Medicine, University of Texas Health San Antonio, Texas
| | - Teresa Whiteside
- University of Pittsburgh and Hillman Comprehensive Cancer Center, Pittsburgh, Pennsylvania
| | - Ilona Kryczek
- Tulane Medical School, Department of Medicine, New Orleans, Louisiana
| | - Weiping Zou
- Tulane Medical School, Department of Medicine, New Orleans, Louisiana
| | - Tyler J Curiel
- The Graduate School of Biomedical Sciences, University of Texas Health San Antonio, Texas. .,Department of Medicine, University of Texas Health San Antonio, Texas.,Mays Cancer Center, University of Texas Health, San Antonio, Texas
| |
Collapse
|
8
|
Chalons P, Courtaut F, Limagne E, Chalmin F, Cantos-Villar E, Richard T, Auger C, Chabert P, Schini-Kerth V, Ghiringhelli F, Aires V, Delmas D. Red Wine Extract Disrupts Th17 Lymphocyte Differentiation in a Colorectal Cancer Context. Mol Nutr Food Res 2020; 64:e1901286. [PMID: 32306526 DOI: 10.1002/mnfr.201901286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/18/2020] [Indexed: 12/13/2022]
Abstract
SCOPE Scope: It is well established that immune response and inflammation promote tumoral progression. Immune cells communicate through direct contact or through cytokine secretion, and it is the pro-inflammatory status that will tip the balance toward tumor progression or anti-tumor immunity. It is demonstrated here that a red wine extract (RWE) can decrease inflammation through its action on the inflammasome complex. This study determines whether an RWE could impact other key actors of inflammation, including T helper 17 (Th17) immune cells in particular. METHODS AND RESULTS Methods and results: Using an RWE containing 4.16 g of polyphenols/liter of wine, it is shown that RWE decreases colorectal cancer cells in vitro and induces a reduction in colorectal tumor growth associated with a decrease in tumor-infiltrating lymphocytes in vivo. The process of T-lymphocyte differentiation in Th17 cells is altered by RWE, as revealed by the decrease in the expression of key actors controlling this process, such as signal transducer and activator of transcription 3 and retinoid acid-related orphan receptor γt. This disruption is associated with an inhibition of inflammatory interleukin 17 secretion. CONCLUSION The data highlights the major involvement of Th17 immune cells in the biological effects of an RWE.
Collapse
Affiliation(s)
- Pauline Chalons
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Flavie Courtaut
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emeric Limagne
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France
| | - Fanny Chalmin
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emma Cantos-Villar
- Instituto de Investigación y Formación Agraria y Pesquera (IFAPA) Rancho de La Merced, Ctra. Trebujena, 11.471 Jerez de la Frontera, Cadiz, Spain
| | - Tristan Richard
- Université de Bordeaux, Unité de Recherche Œnologie, EA 4577, USC 1366 INRA, Equipe Molécules d'Intérêt Biologique - ISVV, Villenave-d'Ornon, F-33882, France
| | - Cyril Auger
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - Philippe Chabert
- UMR CNRS 7021 - Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Illkirch-Graffenstaden, F-67401, France
| | - Valérie Schini-Kerth
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - François Ghiringhelli
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| |
Collapse
|
9
|
Li C, Course MM, McNeish IA, Drescher CW, Valdmanis PN, Lieber A. Prophylactic In Vivo Hematopoietic Stem Cell Gene Therapy with an Immune Checkpoint Inhibitor Reverses Tumor Growth in Syngeneic Mouse Tumor Models. Cancer Res 2020; 80:549-560. [PMID: 31727629 PMCID: PMC7002220 DOI: 10.1158/0008-5472.can-19-1044] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 10/14/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022]
Abstract
Population-wide testing for cancer-associated mutations has established that more than one-fifth of ovarian and breast carcinomas are associated with inherited risk. Salpingo-oophorectomy and/or mastectomy are currently the only effective options offered to women with high-risk germline mutations. Our goal here is to develop a long-lasting approach that provides immunoprophylaxis for mutation carriers. Our approach leverages the fact that at early stages, tumors recruit hematopoietic stem/progenitor cells (HSPC) from the bone marrow and differentiate them into tumor-supporting cells. We developed a technically simple technology to genetically modify HSPCs in vivo. The technology involves HSPC mobilization and intravenous injection of an integrating HDAd5/35++ vector. In vivo HSPC transduction with a GFP-expressing vector and subsequent implantation of syngeneic tumor cells showed >80% GFP marking in tumor-infiltrating leukocytes. To control expression of transgenes, we developed a miRNA regulation system that is activated only when HSPCs are recruited to and differentiated by the tumor. We tested our approach using the immune checkpoint inhibitor anti-PD-L1-γ1 as an effector gene. In in vivo HSPC-transduced mice with implanted mouse mammary carcinoma (MMC) tumors, after initial tumor growth, tumors regressed and did not recur. Conventional treatment with an anti-PD-L1 mAb had no significant antitumor effect, indicating that early, self-activating expression of anti-PD-L1-γ1 can overcome the immunosuppressive environment in MMC tumors. The efficacy and safety of this approach was further validated in an ovarian cancer model with typical germline mutations (ID8 p53-/- brca2-/-), both in a prophylactic and therapeutic setting. This HSPC gene therapy approach has potential for clinical translation. SIGNIFICANCE: Considering the limited prophylactic options that are currently offered to women with high-risk germ-line mutations, the in vivo HSPC gene therapy approach is a promising strategy that addresses a major medical problem.
Collapse
Affiliation(s)
- Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington
| | - Meredith M Course
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington
| | | | | | - Paul N Valdmanis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington.
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
10
|
Hashemi V, Maleki LA, Esmaily M, Masjedi A, Ghalamfarsa G, Namdar A, Yousefi M, Yousefi B, Jadidi-Niaragh F. Regulatory T cells in breast cancer as a potent anti-cancer therapeutic target. Int Immunopharmacol 2019; 78:106087. [PMID: 31841758 DOI: 10.1016/j.intimp.2019.106087] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/23/2019] [Accepted: 11/25/2019] [Indexed: 02/08/2023]
Abstract
Despite marked advances in treatment approaches, breast cancer is still going to be more prevalent, worldwide. High levels of regulatory T (Treg) cells have repeatedly been demonstrated in circulation, lymph nodes, and tumor samples from patients with various cancer types. The transcription factor Forkhead box protein 3 (Foxp3)-expressing Treg cells have the high suppressive potential of the immune system and are fundamental in preserving immune homeostasis and self-tolerance. However, they enhance tumor development by curbing efficient anti-tumor immune mechanisms in malignancies. Moreover, the accumulation of Treg cells in breast tumors is related to the short overall survival of patients. Treg cell frequency has been applied as an independent predicting factor to diagnose patients with a high risk of relapse. Pulling out all populations of Treg cells to promote the efficacy of anticancer treatment methods may potentially lead to hazardous autoimmune disorders. Thus, realizing the exact structure of tumor-infiltrating Treg cells is pivotal to efficiently target Treg cells in tumors. There are exclusive and non-exclusive approaches to lower down and degrade the number/function of Treg cells. These approaches can include inhibiting tumoral migration, depletion, interference with function, and utilizing T cell plasticity. This review article attempts to clarify the implications concerning the involvement of Treg cells in breast cancer progression and discuss the current approaches in the treatment of this cancer via modulation of Treg cells function.
Collapse
Affiliation(s)
- Vida Hashemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Maryam Esmaily
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Masjedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Afshin Namdar
- Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Canada
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Stasikowska-Kanicka O, Wągrowska-Danilewicz M, Danilewicz M. Immunohistochemical Analysis of Foxp3 +, CD4 +, CD8 + Cell Infiltrates and PD-L1 in Oral Squamous Cell Carcinoma. Pathol Oncol Res 2018; 24:497-505. [PMID: 28669079 PMCID: PMC5972165 DOI: 10.1007/s12253-017-0270-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 06/22/2017] [Indexed: 11/26/2022]
Abstract
The immunoexpression of the PD-L1 and the number of immune infiltrating cells have been shown to be a significant prognostic factors in various human cancers. Immunohistochemical method was used to examine the immunoexpression of PD-L1 and number of Foxp3+, CD4+, CD8+ cells in 78 cases of oral squamous cell carcinomas (OSCCs): with better prognosis - OSCCBP (n = 37), and with poorer prognosis - OSCCPP (n = 41), and 18 cases of normal mucosa as a control. The immunoexpression of PD-L1 and the mean number of Foxp3+ cells was significantly increased in OSCCPP group in comparison to OSCCBP and control groups. The mean number of CD4+ cells was significantly increased in OSCCPP group in comparison to OSCCBP and control groups. CD8+ cells were significantly more numerous in OSCCBP group in comparison to OSCCPP and control group. In both OSCCPP and OSCCBP groups there were positive significant correlations between number of Foxp3+ and CD4+ cells. We found positive correlations between the immunoexpression of PD-L1 and numbers of Foxp3+ cells, and negative correlation between the immunoexpression of PD-L1 and numbers of CD8+ cells in both OSCCPP and OSCCBP groups. We found also significant positive correlation between immunoexpression of PD-L1 and the number of CD4+ cells in OSCCPP group. In conclusion, our findings support the hypothesis of involvement of Tregs and PD-L1 in OSCC development and progression.
Collapse
Affiliation(s)
- Olga Stasikowska-Kanicka
- Department of Nephropathology, Medical University of Lodz, ul. Czechoslowacka 8/10, 92-216 Lodz, Poland
| | | | - Marian Danilewicz
- Department of Pathomorphology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
12
|
Rostamzadeh D, Kazemi T, Amirghofran Z, Shabani M. Update on Fc receptor-like (FCRL) family: new immunoregulatory players in health and diseases. Expert Opin Ther Targets 2018; 22:487-502. [PMID: 29737217 DOI: 10.1080/14728222.2018.1472768] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Fc receptor-like (FCRL) molecules, as recently identified members of the immunoglobulin superfamily (IgSF), are preferentially expressed by B-cells. They have variable number of extracellular immunoglobulin-like domains and cytoplasmic activating ITAMs and/or inhibitory ITIMs. FCRL1-5 are dominantly expressed in different stages of B-cells development. But, FCRL6 is preferentially expressed in different subsets of T-cells and NK cells. FCRL1-5 could regulate different features of B-cell evolution such as development, differentiation, activation, antibody secretion and isotype switching. Areas covered: Improved understanding of FCRL expression may grant B-cells and finally its signaling pathways, alone or in cooperation with other signaling molecules, as interesting new targets for diagnostic, monitoring and immunotherapeutic modalities; although further investigations remain to be defined. Recent investigations on different family members of FCRL proteins have substantiated their differential expression on different tissues, malignancies, immune related disease and infectious diseases. Expert opinion: FCRLs restricted expressions in normal B-cells and T-cell subsets accompanied with their overexpression in B-cell malignancies introduce them as logical candidates for the development of antibody- and cell-based immunotherapy approaches in B-cell malignancies, immune-mediated and infectious diseases. FCRLs would be applied as attractive and specific targets for immunodiagnostic approaches, clinical prognosis as well as disease monitoring of relevant patients.
Collapse
Affiliation(s)
- Davood Rostamzadeh
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Tohid Kazemi
- b Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Zahra Amirghofran
- c Department of Immunology, Medical School , Shiraz University of Medical Sciences , Shiraz , Iran.,d Autoimmune Disease Research Center and Medicinal and Natural Products Chemistry Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mahdi Shabani
- e Department of Immunology, School of Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,f Monoclonal Antibody Research Center , Avicenna Research Institute, ACECR , Tehran , Iran
| |
Collapse
|
13
|
O'Konek JJ, Ambrosino E, Bloom AC, Pasquet L, Massilamany C, Xia Z, Terabe M, Berzofsky JA. Differential Regulation of T-cell mediated anti-tumor memory and cross-protection against the same tumor in lungs versus skin. Oncoimmunology 2018; 7:e1439305. [PMID: 29900040 DOI: 10.1080/2162402x.2018.1439305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 01/06/2023] Open
Abstract
A major advantage of immunotherapy of cancer is that effector cells induced at one site should be able to kill metastatic cancer cells in other sites or tissues. However, different tissues have unique immune components, and very little is known about whether effector T cells induced against tumors in one tissue can work against the same tumors in other tissues. Here, we used CT26 murine tumor models to investigate anti-tumor immune responses in the skin and lungs and characterized cross-protection between the two tissues. Blockade of the function of Treg cells with anti-CD25 allowed for T cell-dependent rejection of s.c. tumors. When these mice were simultaneously inoculated i.v. with CT26, they also rejected tumors in the lung. Interestingly, in the absence of s.c. tumors, anti-CD25 treatment alone had no effect on lung tumor growth. These observations suggested that T cell-mediated anti-tumor protective immunity induced against s.c. tumors can also protect against lung metastases of the same tumors. In contrast, NKT cell-deficiency in CD1d-/- mice conferred significant protection against lung tumors but had no effect on the growth of tumors in the skin, and tumor rejection induced against the CT26 in the lung did not confer protection for the same tumor cells in the skin. Thus, effector cells against the same tumor do not work in all tissues, and the induction site of the effector T cells is critical to control metastasis. Further, the regulation of tumor immunity may be different for the same tumor in different anatomical locations.
Collapse
Affiliation(s)
- Jessica J O'Konek
- Vaccine Branch, CCR, NCI, NIH Bethesda, MD USA.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Elena Ambrosino
- Vaccine Branch, CCR, NCI, NIH Bethesda, MD USA.,Institute for Public Health Genomics, Department of Genetics & Cell Biology, School for Oncology & Developmental Biology (GROW), FHML, Maastricht University, The Netherlands
| | | | | | | | - Zheng Xia
- Vaccine Branch, CCR, NCI, NIH Bethesda, MD USA
| | | | | |
Collapse
|
14
|
Nakasone ES, Hurvitz SA, McCann KE. Harnessing the immune system in the battle against breast cancer. Drugs Context 2018; 7:212520. [PMID: 29456568 PMCID: PMC5810622 DOI: 10.7573/dic.212520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most prevalent malignancy in women and the second most common cause of cancer-related death worldwide. Despite major innovations in early detection and advanced therapeutics, up to 30% of women with node-negative breast cancer and 70% of women with node-positive breast cancer will develop recurrence. The recognition that breast tumors are infiltrated by a complex array of immune cells that influence their development, progression, and metastasis, as well as their responsiveness to systemic therapies has sparked major interest in the development of immunotherapies. In fact, not only the native host immune system can be altered to promote potent antitumor response, but also its components can be manipulated to generate effective therapeutic strategies. We present here a review of the major approaches to immunotherapy in breast cancers, both successes and failures, as well as new therapies on the horizon.
Collapse
Affiliation(s)
- Elizabeth S Nakasone
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Sara A Hurvitz
- Division of Hematology/Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kelly E McCann
- Division of Hematology/Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
15
|
Wang F, Wang L, Wu J, Sokirniy I, Nguyen P, Bregnard T, Weinstock J, Mattern M, Bezsonova I, Hancock WW, Kumar S. Active site-targeted covalent irreversible inhibitors of USP7 impair the functions of Foxp3+ T-regulatory cells by promoting ubiquitination of Tip60. PLoS One 2017; 12:e0189744. [PMID: 29236775 PMCID: PMC5728538 DOI: 10.1371/journal.pone.0189744] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022] Open
Abstract
Accumulation of Foxp3+ T-regulatory (Treg) cells in the tumor microenvironment is associated with tumor immune evasion and poor patient outcome in the case of many solid tumors. Current therapeutic strategies for blocking Treg functions are not Treg-specific, and display only modest and transient efficacy. Recent studies revealed that ubiquitin-specific protease 7 (USP7) is essential for Treg functions by stabilizing expression of Tip60 and Foxp3, which together are central to the development and maintenance of the Treg cell lineage. Pharmacological inhibition of USP7 is therefore a promising strategy for suppressing Treg functions and promoting anti-tumor immunity. Previously, we reported the P5091 series of small molecule USP7 inhibitors and demonstrated their direct anti-tumor activity in vivo using xenograft models. However, the precise mechanism of action of these compounds was not well defined. In this study, we report the development and characterization of P217564, a second-generation USP7 inhibitor with improved potency and selectivity. P217564 selectively targets the catalytic cleft of USP7 and modifies its active site cysteine (C223) by forming a covalent adduct. Irreversible inhibition of USP7 results in durable downstream biological responses in cells, including down-regulation of Tip60 and consequent impairment of Treg suppressive function. In addition, we demonstrate that both USP7 and various USP7 substrates are subjected to Lys48-mediated ubiquitin modification, consistent with increased proteasomal degradation of these proteins because of USP7 inhibition.
Collapse
Affiliation(s)
- Feng Wang
- Progenra Inc, Malvern, Pennsylvania, United States of America
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jian Wu
- Progenra Inc, Malvern, Pennsylvania, United States of America
| | - Ivan Sokirniy
- Progenra Inc, Malvern, Pennsylvania, United States of America
| | - Phuong Nguyen
- Progenra Inc, Malvern, Pennsylvania, United States of America
| | - Thomas Bregnard
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, Connecticut, United States of America
| | | | - Michael Mattern
- Progenra Inc, Malvern, Pennsylvania, United States of America
| | - Irina Bezsonova
- Department of Molecular Biology and Biophysics, UCONN Health, Farmington, Connecticut, United States of America
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Suresh Kumar
- Progenra Inc, Malvern, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Lamichhane P, Karyampudi L, Shreeder B, Krempski J, Bahr D, Daum J, Kalli KR, Goode EL, Block MS, Cannon MJ, Knutson KL. IL10 Release upon PD-1 Blockade Sustains Immunosuppression in Ovarian Cancer. Cancer Res 2017; 77:6667-6678. [PMID: 28993412 DOI: 10.1158/0008-5472.can-17-0740] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/24/2017] [Accepted: 09/27/2017] [Indexed: 12/21/2022]
Abstract
Ligation of programmed cell death-1 (PD-1) in the tumor microenvironment is known to inhibit effective adaptive antitumor immunity. Blockade of PD-1 in humans has resulted in impressive, durable regression responses in select tumor types. However, durable responses have been elusive in ovarian cancer patients. PD-1 was recently shown to be expressed on and thereby impair the functions of tumor-infiltrating murine and human myeloid dendritic cells (TIDC) in ovarian cancer. In the present work, we characterize the regulation of PD-1 expression and the effects of PD-1 blockade on TIDC. Treatment of TIDC and bone marrow-derived dendritic cells (DC) with IL10 led to increased PD-1 expression. Both groups of DCs also responded to PD-1 blockade by increasing production of IL10. Similarly, treatment of ovarian tumor-bearing mice with PD-1 blocking antibody resulted in an increase in IL10 levels in both serum and ascites. While PD-1 blockade or IL10 neutralization as monotherapies were inefficient, combination of these two led to improved survival and delayed tumor growth; this was accompanied by augmented antitumor T- and B-cell responses and decreased infiltration of immunosuppressive MDSC. Taken together, our findings implicate compensatory release of IL10 as one of the adaptive resistance mechanisms that undermine the efficacy of anti-PD-1 (or anti-PD-L1) monotherapies and prompt further studies aimed at identifying such resistance mechanisms. Cancer Res; 77(23); 6667-78. ©2017 AACR.
Collapse
Affiliation(s)
- Purushottam Lamichhane
- Department of Immunology, Mayo Clinic Rochester, Minnesota.,Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - Lavakumar Karyampudi
- Department of Immunology, Mayo Clinic Rochester, Minnesota.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - Barath Shreeder
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - James Krempski
- Department of Immunology, Mayo Clinic Rochester, Minnesota
| | - Deborah Bahr
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida
| | - Joshua Daum
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida
| | | | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic Rochester, Minnesota
| | | | - Martin J Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic Rochester, Minnesota. .,Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| |
Collapse
|
17
|
Bahrami A, Hassanian SM, Khazaei M, Hasanzadeh M, Shahidsales S, Maftouh M, Ferns GA, Avan A. The Therapeutic Potential of Targeting Tumor Microenvironment in Breast Cancer: Rational Strategies and Recent Progress. J Cell Biochem 2017; 119:111-122. [DOI: 10.1002/jcb.26183] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/01/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Medical Biochemistry, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Majid Khazaei
- Department of Physiology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - Mina Maftouh
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationFalmer, BrightonSussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Cancer Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
18
|
Majidinia M, Yousefi B. Breast tumor stroma: A driving force in the development of resistance to therapies. Chem Biol Drug Des 2017; 89:309-318. [PMID: 28042683 DOI: 10.1111/cbdd.12893] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 09/15/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most common cancer and the second leading cause of cancer-related death in women worldwide. In spite of huge advancements in early detection and ever-increasing knowledge of breast cancer biology, approximately 30% of patients with early-stage breast cancer experience disease recurrence. Most patients are chemosensitive and cancer free immediately after the treatment. About 50% to 70% of breast cancer patients, however, will relapse within 1 year. Such a relapse is usually concomitant with adenocarcinoma cells acquiring a chemoresistant phenotype. Both de novo and acquired chemoresistance are poorly understood and present a major burden in the treatment of breast cancer. Although, previously, chemoresistance was largely linked to genetic alterations within the cancer cells, recent investigations are indicating that chemoresistance can also be associated with the tumor microenvironment. Nowadays, it is widely believed that tumor microenvironment is a key player in tumor progression and response to treatment. In this study, we will review the interactions of breast tumor cells with their microenvironment, present the latest research on the resistance mediated by the stromal component in breast cancer, and discuss the potential therapeutic strategies that can be exploited to treat breast cancers by targeting tumor microenvironment.
Collapse
Affiliation(s)
- Maryam Majidinia
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University Medical Sciences, Urmia, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Wang L, Kumar S, Dahiya S, Wang F, Wu J, Newick K, Han R, Samanta A, Beier UH, Akimova T, Bhatti TR, Nicholson B, Kodrasov MP, Agarwal S, Sterner DE, Gu W, Weinstock J, Butt TR, Albelda SM, Hancock WW. Ubiquitin-specific Protease-7 Inhibition Impairs Tip60-dependent Foxp3+ T-regulatory Cell Function and Promotes Antitumor Immunity. EBioMedicine 2016; 13:99-112. [PMID: 27769803 PMCID: PMC5264272 DOI: 10.1016/j.ebiom.2016.10.018] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 02/08/2023] Open
Abstract
Foxp3 + T-regulatory (Treg) cells are known to suppress protective host immune responses to a wide variety of solid tumors, but their therapeutic targeting is largely restricted to their transient depletion or “secondary” modulation, e.g. using anti-CTLA-4 monoclonal antibody. Our ongoing studies of the post-translational modifications that regulate Foxp3 demonstrated that the histone/protein acetyltransferase, Tip60, plays a dominant role in promoting acetylation, dimerization and function in Treg cells. We now show that the ubiquitin-specific protease, Usp7, controls Treg function largely by stabilizing the expression and promoting the multimerization of Tip60 and Foxp3. Genetic or pharmacologic targeting of Usp7 impairs Foxp3 + Treg suppressive functions, while conventional T cell responses remain intact. As a result, pharmacologic inhibitors of Usp7 can limit tumor growth in immunocompetent mice, and promote the efficacy of antitumor vaccines and immune checkpoint therapy with anti-PD1 monoclonal antibody in murine models. Hence, pharmacologic therapy with Usp7 inhibitors may have an important role in future cancer immunotherapy. Conditional deletion of Usp7 in Foxp3 + Treg cells causes rapidly lethal autoimmunity.
Pharmacologic inhibition of Usp7 impairs Treg but not conventional T cell function.
Usp7 targeting alone, or in conjunction with other therapies, promotes anti-tumor immunity.
T-regulatory (Treg) cells are essential to regulation of the immune system, and are characterized by their expression of the transcription factor, Foxp3. Foxp3 is subject to ubiquitination and degradation via the proteasome. We now show that the deubiquitinase, Usp7, is a key regulator of Foxp3 + Treg biology through controlling levels of the histone acetyltransferase, Tip60 and, to a lesser extent, Foxp3. Gene deletion or pharmacologic inhibition of Usp7 impairs Treg but not conventional T cell functions. The pharmacologic targeting of Usp7 alone, or in conjunction with additional therapeutic strategies, is of significant benefit in promoting host anti-tumor immunity.
Collapse
Affiliation(s)
- Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Satinder Dahiya
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feng Wang
- Progenra, Inc., Malvern, PA 19355, USA
| | - Jian Wu
- Progenra, Inc., Malvern, PA 19355, USA
| | - Kheng Newick
- Pulmonary, Allergy & Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA19104, USA
| | - Rongxiang Han
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arabinda Samanta
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ulf H Beier
- Division of Nephrology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA19104, USA
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tricia R Bhatti
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | - Wei Gu
- Institute for Cancer Genetics and Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | - Steven M Albelda
- Pulmonary, Allergy & Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA19104, USA
| | - Wayne W Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Chimeric Antigen Receptor-Modified T Cells for Solid Tumors: Challenges and Prospects. J Immunol Res 2016; 2016:3850839. [PMID: 26998495 PMCID: PMC4779545 DOI: 10.1155/2016/3850839] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/18/2015] [Accepted: 01/28/2016] [Indexed: 12/31/2022] Open
Abstract
Recent studies have highlighted the successes of chimeric antigen receptor-modified T- (CART-) cell-based therapy for B-cell malignancies, and early phase clinical trials have been launched in recent years. The few published clinical studies of CART cells in solid tumors have addressed safety and feasibility, but the clinical outcome data are limited. Although antitumor effects were confirmed in vitro and in animal models, CART-cell-based therapy still faces several challenges when directed towards solid tumors, and it has been difficult to achieve the desired outcomes in clinical practice. Many studies have struggled to improve the clinical responses to and benefits of CART-cell treatment of solid tumors. In this review, the status quo of CART cells and their clinical applications for solid tumors will be summarized first. Importantly, we will suggest improvements that could increase the therapeutic effectiveness of CART cells for solid tumors and their future clinical applications. These interventions will make treatment with CART cells an effective and routine therapy for solid tumors.
Collapse
|
21
|
Karyampudi L, Lamichhane P, Krempski J, Kalli KR, Behrens MD, Vargas DM, Hartmann LC, Janco JMT, Dong H, Hedin KE, Dietz AB, Goode EL, Knutson KL. PD-1 Blunts the Function of Ovarian Tumor-Infiltrating Dendritic Cells by Inactivating NF-κB. Cancer Res 2015; 76:239-50. [PMID: 26567141 DOI: 10.1158/0008-5472.can-15-0748] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 10/13/2015] [Indexed: 01/22/2023]
Abstract
The PD-1:PD-L1 immune signaling axis mediates suppression of T-cell-dependent tumor immunity. PD-1 expression was recently found to be upregulated on tumor-infiltrating murine (CD11c(+)CD11b(+)CD8(-)CD209a(+)) and human (CD1c(+)CD19(-)) myeloid dendritic cells (TIDC), an innate immune cell type also implicated in immune escape. However, there is little knowledge concerning how PD-1 regulates innate immune cells. In this study, we examined the role of PD-1 in TIDCs derived from mice bearing ovarian tumors. Similar to lymphocytes, TIDC expression of PD-1 was associated with expression of the adapter protein SHP-2, which signals to NF-κB; however, in contrast to its role in lymphocytes, we found that expression of PD-1 in TIDC tonically paralyzed NF-κB activation. Further mechanistic investigations showed that PD-1 blocked NF-κB-dependent cytokine release in a SHP-2-dependent manner. Conversely, inhibition of NF-κB-mediated antigen presentation by PD-1 occurred independently of SHP-2. Collectively, our findings revealed that PD-1 acts in a distinct manner in innate immune cells compared with adaptive immune cells, prompting further investigations of the signaling pathways controlled by this central mediator of immune escape in cancer.
Collapse
Affiliation(s)
| | - Purushottam Lamichhane
- Vaccine and Gene Therapy Institute, Port St. Lucie, Florida. Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - James Krempski
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Doris M Vargas
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | | | - Jo Marie T Janco
- Department of Gynecologic Surgery Mayo Clinic, Mayo Clinic, Rochester, Minnesota
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Karen E Hedin
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Allan B Dietz
- Human Cell Therapy Lab, Mayo Clinic, Rochester, Minnesota
| | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Keith L Knutson
- Vaccine and Gene Therapy Institute, Port St. Lucie, Florida. Department of Immunology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
22
|
Regulatory T cells, inherited variation, and clinical outcome in epithelial ovarian cancer. Cancer Immunol Immunother 2015; 64:1495-504. [PMID: 26298430 DOI: 10.1007/s00262-015-1753-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 08/12/2015] [Indexed: 02/08/2023]
Abstract
The immune system constitutes one of the host factors modifying outcomes in ovarian cancer. Regulatory T cells (Tregs) are believed to be a major factor in preventing the immune response from destroying ovarian cancers. Understanding mechanisms that regulate Tregs in the tumor microenvironment could lead to the identification of novel targets aimed at reducing their influence. In this study, we used immunofluorescence-based microscopy to enumerate Tregs, total CD4 T cells, and CD8(+) cytotoxic T cells in fresh frozen tumors from over 400 patients with ovarian cancer (>80 % high-grade serous). We sought to determine whether Tregs were associated with survival and genetic variation in 79 genes known to influence Treg induction, trafficking, or function. We used Cox regression, accounting for known prognostic factors, to estimate hazard ratios (HRs) associated with T cell counts and ratios. We found that the ratios of CD8 T cells and total CD4 T cells to Tregs were associated with improved overall survival (CD8/Treg HR 0.84, p = 0.0089; CD4/Treg HR 0.88, p = 0.046) and with genetic variation in IL-10 (p = 0.0073 and 0.01, respectively). In multivariate analyses, the associations between the ratios and overall survival remained similar (IL-10 and clinical covariate-adjusted CD8/Treg HR 0.85, p = 0.031; CD4/Treg HR 0.87, p = 0.093), suggesting that this association was not driven by variation in IL-10. Thus, integration of novel tumor phenotyping measures with extensive clinical and genetic information suggests that the ratio of T cells to Tregs may be prognostic of outcome in ovarian cancer, regardless of inherited genotype in genes related to Tregs.
Collapse
|
23
|
Heon EK, Wulan H, Macdonald LP, Malek AO, Braunstein GH, Eaves CG, Schattner MD, Allen PM, Alexander MO, Hawkins CA, McGovern DW, Freeman RL, Amir EP, Huse JD, Zaltzman JS, Kauff NP, Meyers PG, Gleason MH, Overholtzer MG, Wiseman SS, Streutker CD, Asa SW, McAlindon TP, Newcomb PO, Sorensen PM, Press OA. IL-15 induces strong but short-lived tumor-infiltrating CD8 T cell responses through the regulation of Tim-3 in breast cancer. Biochem Biophys Res Commun 2015; 464:360-6. [PMID: 26141233 DOI: 10.1016/j.bbrc.2015.06.162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 06/26/2015] [Indexed: 01/21/2023]
Abstract
IL-15 has pivotal roles in the control of CD8(+) memory T cells and has been investigated as a therapeutic option in cancer therapy. Although IL-15 and IL-2 share many functions together, including the stimulation of CD8 T cell proliferation and IFN-γ production, the different in vivo roles of IL-15 and IL-2 have been increasingly recognized. Here, we explored the different effects of IL-15 and IL-2 on tumor-infiltrating (TI) T cells from resected breast tumors. We found that neither IL-2 nor IL-15 induced intratumoral CD8 T cell proliferation by itself, but after CD3/CD28-stimulation, IL-15 induced significantly higher proliferation than IL-2 during early time points, at day 2, day 3 and day 6. However, the IL-15-induced proliferation leveled off at day 9 and day 12, whereas IL-2 induced lower but progressive proliferation at each time point. Furthermore, IL-15 caused an early and robust increase of IFN-γ in the supernatant of TI cell cultures, which diminished at later time points, while the IL-2-induced IFN-γ production remained constant over time. In addition, the IL-15-costimulated CD8 T cells presented higher frequencies of apoptotic cells. The diminishing IL-15-induced response was possibly due to regulatory and/or exhaustion mechanisms. We did not observe increased IL-10 or PD-1 upregulation, but we have found an increase of Tim-3 upregulation on IL-15-, but not IL-2-stimulated cells. Blocking Tim-3 function using anti-Tim-3 antibodies resulted in increased IL-15-induced proliferation and IFN-γ production for a prolonged period of time, whereas adding Tim-3 ligand galectin 9 led to reduced proliferation and IFN-γ production. Our results suggest that IL-15 in combination of Tim-3 blocking antibodies could potentially act as an IL-2 alternative in tumor CD8 T cell expansion in vitro, a crucial step in adoptive T cell therapy.
Collapse
Affiliation(s)
- Elise K Heon
- University of Maryland Medical Center, Baltimore, MD 21201, United States
| | - Hasi Wulan
- Department of Plastic and Reconstructive Surgery, PLA General Hospital, Beijing, 100853, China
| | | | - Adel O Malek
- Brown University, Providence, RI 02912, United States
| | | | | | | | - Peter M Allen
- University of Wisconsin, Madison, WI 53706, United States
| | | | | | | | | | - Eitan P Amir
- University of Illinois, Chicago, IL 60607, United States
| | - Jason D Huse
- University of Illinois, Chicago, IL 60607, United States
| | | | - Noah P Kauff
- University of Texas, Austin, TX 78712, United States
| | - Paul G Meyers
- University of Texas, Austin, TX 78712, United States
| | | | | | - Sam S Wiseman
- Ohio State University, Columbus, OH 43210, United States
| | | | - Sylvia W Asa
- Ohio State University, Columbus, OH 43210, United States
| | | | | | | | - Oliver A Press
- University of Illinois, Chicago, IL 60607, United States.
| |
Collapse
|
24
|
Kravchenko J, Corsini E, Williams MA, Decker W, Manjili MH, Otsuki T, Singh N, Al-Mulla F, Al-Temaimi R, Amedei A, Colacci AM, Vaccari M, Mondello C, Scovassi AI, Raju J, Hamid RA, Memeo L, Forte S, Roy R, Woodrick J, Salem HK, Ryan EP, Brown DG, Bisson WH, Lowe L, Lyerly HK. Chemical compounds from anthropogenic environment and immune evasion mechanisms: potential interactions. Carcinogenesis 2015; 36 Suppl 1:S111-27. [PMID: 26002081 DOI: 10.1093/carcin/bgv033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 01/19/2015] [Indexed: 02/07/2023] Open
Abstract
An increasing number of studies suggest an important role of host immunity as a barrier to tumor formation and progression. Complex mechanisms and multiple pathways are involved in evading innate and adaptive immune responses, with a broad spectrum of chemicals displaying the potential to adversely influence immunosurveillance. The evaluation of the cumulative effects of low-dose exposures from the occupational and natural environment, especially if multiple chemicals target the same gene(s) or pathway(s), is a challenge. We reviewed common environmental chemicals and discussed their potential effects on immunosurveillance. Our overarching objective was to review related signaling pathways influencing immune surveillance such as the pathways involving PI3K/Akt, chemokines, TGF-β, FAK, IGF-1, HIF-1α, IL-6, IL-1α, CTLA-4 and PD-1/PDL-1 could individually or collectively impact immunosurveillance. A number of chemicals that are common in the anthropogenic environment such as fungicides (maneb, fluoxastrobin and pyroclostrobin), herbicides (atrazine), insecticides (pyridaben and azamethiphos), the components of personal care products (triclosan and bisphenol A) and diethylhexylphthalate with pathways critical to tumor immunosurveillance. At this time, these chemicals are not recognized as human carcinogens; however, it is known that they these chemicalscan simultaneously persist in the environment and appear to have some potential interfere with the host immune response, therefore potentially contributing to promotion interacting with of immune evasion mechanisms, and promoting subsequent tumor growth and progression.
Collapse
Affiliation(s)
- Julia Kravchenko
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
| | - Emanuela Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, School of Pharmacy, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marc A Williams
- MEDCOM Army Institute of Public Health, Toxicology Portfolio - Health Effects Research Program, Aberdeen Proving Ground, Edgewood, Baltimore, MD 21010, USA
| | - William Decker
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masoud H Manjili
- Department of Microbiology and Immunology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Takemi Otsuki
- Department of Hygiene, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Faha Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hosni K Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO, 80523-1680, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO, 80523-1680, USA
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA,
| | - Leroy Lowe
- Getting to Know Cancer, Nova Scotia, Canada and
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
25
|
Wei M, Marino J, Trowell A, Zhang H, Stromp Peraino J, Rajasekera PV, Madsen JC, Sachs DH, Huang CA, Benichou G, Wang Z. Diphtheria toxin-based recombinant murine IL-2 fusion toxin for depleting murine regulatory T cells in vivo. Protein Eng Des Sel 2015; 27:289-95. [PMID: 25147093 DOI: 10.1093/protein/gzu034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Regulatory T cells (Tregs) are a subpopulation of CD4(+) T cells which suppress immune responses of effector cells and are known to play a very important role in protection against autoimmune disease development, induction of transplantation tolerance and suppression of effective immune response against tumor cells. An effective in vivo Treg depletion agent would facilitate Treg-associated studies across many research areas. In this study, we have developed diphtheria toxin-based monovalent and bivalent murine IL-2 fusion toxins for depleting murine IL-2 receptor positive cells including CD25(+) Treg in vivo. Their potencies were assessed by in vitro protein synthesis inhibition and cell proliferation inhibition assays using a murine CD25(+) CTLL-2 cell line. Surprisingly, in contrast to our previously developed recombinant fusion toxins, the monovalent isoform (DT390-mIL-2) was approximately 4-fold more potent than its bivalent counterpart (DT390-bi-mIL-2). Binding analysis by flow cytometry demonstrated that the monovalent isoform bound stronger than the bivalent version. In vivo Treg depletion with the monovalent murine IL-2 fusion toxin was performed using C57BL/6J (B6) mice. Spleen Treg were significantly depleted with a maximum reduction of ∼70% and detectable as early as 12 h after the last injection. The spleen Treg numbers were reduced until Day 3 and returned to control levels by Day 7. We believe that this monovalent murine IL-2 fusion toxin will be an effective in vivo murine Treg depleter.
Collapse
Affiliation(s)
- Min Wei
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Jose Marino
- Transplant Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron Trowell
- Transplant Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiping Zhang
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Jaclyn Stromp Peraino
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Priyani V Rajasekera
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Joren C Madsen
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA Transplant Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David H Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Christene A Huang
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| | - Gilles Benichou
- Transplant Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhirui Wang
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA MGH-DF/HCC Recombinant Protein Expression and Purification Core, Boston, MA, USA
| |
Collapse
|
26
|
Zhu S, Lin J, Qiao G, Xu Y, Zou H. Differential regulation and function of tumor-infiltrating T cells in different stages of breast cancer patients. Tumour Biol 2015; 36:7907-13. [DOI: 10.1007/s13277-015-3507-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
|
27
|
Martinez LM, Labovsky V, de Luján Calcagno M, Davies KM, Rivello HG, Bianchi MS, Wernicke A, Vallone VBF, Chasseing NA. CD105 expression on CD34-negative spindle-shaped stromal cells of primary tumor is an unfavorable prognostic marker in early breast cancer patients. PLoS One 2015; 10:e0121421. [PMID: 25803686 PMCID: PMC4372565 DOI: 10.1371/journal.pone.0121421] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 02/12/2015] [Indexed: 02/06/2023] Open
Abstract
Several studies have confirmed that the breast tumor microenvironment drives cancer progression and metastatic development. The aim of our research was to investigate the prognostic significance of the breast tumor microenvironment in untreated early breast cancer patients. Therefore, we analyzed the association of the expression of α-SMA, FSP, CD105 and CD146 in CD34-negative spindle-shaped stromal cells, not associated with the vasculature, in primary breast tumors with classical prognostic marker levels, metastatic recurrence, local relapse, disease-free survival, metastasis-free survival and the overall survival of patients. In the same way, we evaluated the association of the amount of intra-tumor stroma, fibroblasts, collagen deposition, lymphocytic infiltration and myxoid changes in these samples with the clinical-pathological data previously described. This study is the first to demonstrate the high CD105 expression in this stromal cell type as a possible independent marker of unfavorable prognosis in early breast cancer patients. Our study suggests that this new finding can be useful prognostic marker in the clinical-pathological routine.
Collapse
Affiliation(s)
- Leandro Marcelo Martinez
- Immunohematology Laboratory, Experimental Biology and Medicine Institute (IBYME)—National Council of Scientific and Technical Research (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Immunohematology Laboratory, Experimental Biology and Medicine Institute (IBYME)—National Council of Scientific and Technical Research (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María de Luján Calcagno
- Department of Biostatistics, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Kevin Mauro Davies
- Department of Pathological Anatomy, Italian Hospital, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Hernán Garcia Rivello
- Department of Pathological Anatomy, Italian Hospital, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Maria Silvia Bianchi
- Neuroendocrinology Laboratory, Experimental Biology and Medicine Institute (IBYME)—National Council of Scientific and Technical Research (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Wernicke
- Department of Pathological Anatomy, Italian Hospital, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Beatriz Fernández Vallone
- Immunohematology Laboratory, Experimental Biology and Medicine Institute (IBYME)—National Council of Scientific and Technical Research (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Immunohematology Laboratory, Experimental Biology and Medicine Institute (IBYME)—National Council of Scientific and Technical Research (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
28
|
Wertel I, Surówka J, Polak G, Barczyński B, Bednarek W, Jakubowicz-Gil J, Bojarska-Junak A, Kotarski J. Macrophage-derived chemokine CCL22 and regulatory T cells in ovarian cancer patients. Tumour Biol 2015; 36:4811-7. [PMID: 25647263 PMCID: PMC4529457 DOI: 10.1007/s13277-015-3133-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/19/2015] [Indexed: 12/02/2022] Open
Abstract
The study was undertaken to evaluate macrophage-derived chemokine (CCL22) levels in the peritoneal fluid (PF) and plasma of patients with ovarian cancer (n = 93) in relation to regulatory T cells (Tregs; n = 75). The peritoneal fluid CCL22 concentrations were significantly higher in epithelial ovarian cancer (EOC) patients than in patients with benign tumors-serous cystadenoma (n = 32). There was no difference in plasma levels of CCL22 in EOC patients compared with the non-cancer and healthy volunteers (n = 10). There were no significant differences in the plasma and PF CCL22 levels based on tumor grade. However, women with stage IV FIGO (International Federation of Gynecologists and Obstetricians) had significantly higher plasma CCL22 levels than patients with stages I and III. Women with stage I FIGO had significantly higher PF CCL22 levels than patients with stages II and III. Women with endometrioid cystadenocarcinoma had higher PF CCL22 levels than women with undifferentiated carcinoma. The percentage of tumor-infiltrating Tregs (11.06 %) was significantly higher compared to PF (3.05 %) and peripheral blood (PB) (2.01 %). Moreover, the percentage of Tregs was higher in the PF than in the PB of EOC patients. There were no significant differences in the PB, PF, and tumor-infiltrating Tregs percentage based on tumor stage, grade, or histology. Elevated levels of CCL22 found in the ascites could create a chemokine gradient aiding in Treg cells migration. Increased Tregs percentage in the local microenvironment of ovarian cancer might be an important mechanism of immunosuppression.
Collapse
Affiliation(s)
- I Wertel
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland,
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Gad E, Rastetter L, Slota M, Koehnlein M, Treuting PM, Dang Y, Stanton S, Disis ML. Natural history of tumor growth and immune modulation in common spontaneous murine mammary tumor models. Breast Cancer Res Treat 2014; 148:501-10. [PMID: 25395320 PMCID: PMC4280094 DOI: 10.1007/s10549-014-3199-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/04/2014] [Indexed: 12/20/2022]
Abstract
Recent studies in patients with breast cancer suggest the immune microenvironment influences response to therapy. We aimed to evaluate the relationship between growth rates of tumors in common spontaneous mammary tumor models and immune biomarkers evaluated in the tumor and blood. TgMMTV-neu and C3(1)-Tag transgenic mice were followed longitudinally from birth, and MPA-DMBA-treated mice from the time of carcinogen administration, for the development of mammary tumors. Tumor-infiltrating CD4(+) and CD8(+) T-cells, FOXP3(+) T-regulatory cells, and myeloid-derived suppressor cells were assessed by flow cytometry. Serum cytokines were evaluated in subsets of mice. Fine needle aspirates of tumors were collected and RNA was isolated to determine levels of immune and proliferation markers. Age of tumor onset and kinetics of tumor growth were significantly different among the models. Mammary tumors from TgMMTV-neu contained a lower CD8/CD4 ratio than that of other models (p < 0.05). MPA-DMBA-induced tumors contained a higher percentage of FOXP3(+) CD4(+) T-cells (p < 0.01) and MDSC (p < 0.001) compared with the other models. Individuals with significantly slower tumor growth demonstrated higher levels of Type I serum cytokines prior to the development of lesions compared to those with rapid tumor growth. Moreover, the tumors of animals with more rapid tumor growth demonstrated a significant increase in the expression of genes associated with Type II immunity than those with slower-progressing tumors. These data provide a foundation for the development of in vivo models to explore the relationship between endogenous immunity and response to standard therapies for breast cancer.
Collapse
Affiliation(s)
- Ekram Gad
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Lauren Rastetter
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Meredith Slota
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Marlese Koehnlein
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Piper M. Treuting
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Sasha Stanton
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| | - Mary L. Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women’s Health, University of Washington, Seattle, WA
| |
Collapse
|
30
|
Bellone S, Pecorelli S, Cannon MJ, Santin AD. Advances in dendritic cell-based therapeutic vaccines for cervical cancer. Expert Rev Anticancer Ther 2014; 7:1473-86. [DOI: 10.1586/14737140.7.10.1473] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Pietersz GA. Enhanced immune responses to an adenovirus CEA vaccine in CD4+CD25+regulatory T-cell inactivated mice. Expert Rev Vaccines 2014; 6:21-3. [PMID: 17280474 DOI: 10.1586/14760584.6.1.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Geoffrey A Pietersz
- Bio-organic & Medicinal Chemistry Laboratory, Burnet Institute at Austin, Studley Road, Heidelberg, Victoria, 3084, Australia.
| |
Collapse
|
32
|
Mao Y, Keller ET, Garfield DH, Shen K, Wang J. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev 2013; 32:303-15. [PMID: 23114846 DOI: 10.1007/s10555-012-9415-3] [Citation(s) in RCA: 497] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer is a systemic disease encompassing multiple components of both tumor cells themselves and host stromal cells. It is now clear that stromal cells in the tumor microenvironment play an important role in cancer development. Molecular events through which reactive stromal cells affect cancer cells can be defined so that biomarkers and therapeutic targets can be identified. Cancer-associated fibroblasts (CAFs) make up the bulk of cancer stroma and affect the tumor microenvironment such that they promote cancer initiation, angiogenesis, invasion, and metastasis. In breast cancer, CAFs not only promote tumor progression but also induce therapeutic resistance. Accordingly, targeting CAFs provides a novel way to control tumors with therapeutic resistance. This review summarizes the current understandings of tumor stroma in breast cancer with a particular emphasis on the role of CAFs and the therapeutic implications of CAFs. In addition, the effects of other stromal components such as endothelial cells, macrophages, and adipocytes in breast cancer are also discussed. Finally, we describe the biologic markers to categorize patients into a specific and confirmed subtype for personalized treatment.
Collapse
Affiliation(s)
- Yan Mao
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, China
| | | | | | | | | |
Collapse
|
33
|
Preston CC, Maurer MJ, Oberg AL, Visscher DW, Kalli KR, Hartmann LC, Goode EL, Knutson KL. The ratios of CD8+ T cells to CD4+CD25+ FOXP3+ and FOXP3- T cells correlate with poor clinical outcome in human serous ovarian cancer. PLoS One 2013; 8:e80063. [PMID: 24244610 PMCID: PMC3828213 DOI: 10.1371/journal.pone.0080063] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/08/2013] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer is an immune reactive malignancy with a complex immune suppressive network that blunts successful immune eradication. This suppressive microenvironment may be mediated by recruitment or induction of CD4+ regulatory T cells (Tregs). Our study sought to investigate the association of tumor-infiltrating CD4+CD25+FOXP3+ Tregs, and other immune factors, with clinical outcome in serous ovarian cancer patients. We performed immunofluorescence and quantification of intraepithelial tumor-infiltrating triple positive Tregs (CD4+CD25+FOXP3+), as well as CD4+CD25+FOXP3-, CD3+ and CD8+ T cells in tumor specimens from 52 patients with high stage serous ovarian carcinoma. Thirty-one of the patients had good survival (i.e. > 60 months) and 21 had poor survival of < 18 months. Total cell counts as well as cell ratios were compared among these two outcome groups. The total numbers of CD4+CD25+FOXP3+ Tregs, CD4+CD25+FOXP3-, CD3+ and CD8+ cells were not significantly different between the groups. However, higher ratios of CD8+/CD4+CD25+FOXP3+ Treg, CD8+/CD4+ and CD8/CD4+CD25+FOXP3- cells were seen in the good outcome group when compared to the patients with poor outcome. These data show for the first time that the ratios of CD8+ to both CD4+CD25+FOXP3+ Tregs and CD4+CD25+FOXP3- T cells are associated with disease outcome in ovarian cancer. The association being apparent in ratios rather than absolute count of T cells suggests that the effector/suppressor ratio may be a more important indicator of outcome than individual cell count. Thus, immunotherapy strategies that modify the ratio of CD4+CD25+FOXP3+ Tregs or CD4+CD25+FOXP3- T cells to CD8+ effector cells may be useful in improving outcomes in ovarian cancer.
Collapse
Affiliation(s)
- Claudia C. Preston
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthew J. Maurer
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ann L. Oberg
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Daniel W. Visscher
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kimberly R. Kalli
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Lynn C. Hartmann
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ellen L. Goode
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (ELG); (KLK)
| | - Keith L. Knutson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Vaccine and Gene Therapy Institute, Port St. Lucie, Florida, United States of America
- * E-mail: (ELG); (KLK)
| |
Collapse
|
34
|
Izhak L, Ambrosino E, Kato S, Parish ST, O’Konek JJ, Weber H, Xia Z, Venzon D, Berzofsky JA, Terabe M. Delicate balance among three types of T cells in concurrent regulation of tumor immunity. Cancer Res 2013; 73:1514-23. [PMID: 23319803 PMCID: PMC3622595 DOI: 10.1158/0008-5472.can-12-2567] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The nature of the regulatory cell types that dominate in any given tumor is not understood at present. Here, we addressed this question for regulatory T cells (Treg) and type II natural killer T (NKT) cells in syngeneic models of colorectal and renal cancer. In mice with both type I and II NKT cells, or in mice with neither type of NKT cell, Treg depletion was sufficient to protect against tumor outgrowth. Surprisingly, in mice lacking only type I NKT cells, Treg blockade was insufficient for protection. Thus, we hypothesized that type II NKT cells may be neutralized by type I NKT cells, leaving Tregs as the primary suppressor, whereas in mice lacking type I NKT cells, unopposed type II NKT cells could suppress tumor immunity even when Tregs were blocked. We confirmed this hypothesis in 3 ways by reconstituting type I NKT cells as well as selectively blocking or activating type II NKT cells with antibody or the agonist sulfatide, respectively. In this manner, we showed that blockade of both type II NKT cells and Tregs is necessary to abrogate suppression of tumor immunity, but a third cell, the type I NKT cell, determines the balance between these regulatory mechanisms. As patients with cancer often have deficient type I NKT cell function, managing this delicate balance among 3 T-cell subsets may be critical for the success of immunotherapy for human cancer.
Collapse
Affiliation(s)
- Liat Izhak
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Elena Ambrosino
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Stanley T. Parish
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jessica J. O’Konek
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Hannah Weber
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - David Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| |
Collapse
|
35
|
Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y. Higher frequencies of GARP(+)CTLA-4(+)Foxp3(+) T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res 2013; 73:2435-44. [PMID: 23423978 DOI: 10.1158/0008-5472.can-12-3381] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The extent to which T-cell-mediated immune surveillance is impaired in human cancer remains a question of major importance, given its potential impact on the development of generalized treatments of advanced disease where the highest degree of heterogeneity exists. Here, we report the first global analysis of immune dysfunction in patients with advanced hepatocellular carcinoma (HCC). Using multi-parameter fluorescence-activated cell sorting analysis, we quantified the cumulative frequency of regulatory T cells (Treg), exhausted CD4(+) helper T cells, and myeloid-derived suppressor cells (MDSC) to gain concurrent views on the overall level of immune dysfunction in these inoperable patients. We documented augmented numbers of Tregs, MDSC, PD-1(+)-exhausted T cells, and increased levels of immunosuppressive cytokines in patients with HCC, compared with normal controls, revealing a network of potential mechanisms of immune dysregulation in patients with HCC. In dampening T-cell-mediated antitumor immunity, we hypothesized that these processes may facilitate HCC progression and thwart the efficacy of immunotherapeutic interventions. In testing this hypothesis, we showed that combined regimens to deplete Tregs, MDSC, and PD-1(+) T cells in patients with advanced HCC restored production of granzyme B by CD8(+) T cells, reaching levels observed in normal controls and also modestly increased the number of IFN-γ producing CD4(+) T cells. These clinical findings encourage efforts to restore T-cell function in patients with advanced stage disease by highlighting combined approaches to deplete endogenous suppressor cell populations that can also expand effector T-cell populations.
Collapse
Affiliation(s)
- Suresh Kalathil
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | |
Collapse
|
36
|
Foulds GA, Radons J, Kreuzer M, Multhoff G, Pockley AG. Influence of tumors on protective anti-tumor immunity and the effects of irradiation. Front Oncol 2013; 3:14. [PMID: 23378947 PMCID: PMC3561630 DOI: 10.3389/fonc.2013.00014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/15/2013] [Indexed: 12/20/2022] Open
Abstract
Innate and adaptive immunity plays important roles in the development and progression of cancer and it is becoming apparent that tumors can influence the induction of potentially protective responses in a number of ways. The prevalence of immunoregulatory T cell populations in the circulation and tumors of patients with cancer is increased and the presence of these cells appears to present a major barrier to the induction of tumor immunity. One aspect of tumor-mediated immunoregulation which has received comparatively little attention is that which is directed toward natural killer (NK) cells, although evidence that the phenotype and function of NK cell populations are modified in patients with cancer is accumulating. Although the precise mechanisms underlying these localized and systemic immunoregulatory effects remain unclear, tumor-derived factors appear, in part at least, to be involved. The effects could be manifested by an altered function and/or via an influence on the migratory properties of individual cell subsets. A better insight into endogenous immunoregulatory mechanisms and the capacity of tumors to modify the phenotype and function of innate and adaptive immune cells might assist the development of new immunotherapeutic approaches and improve the management of patients with cancer. This article reviews current knowledge relating to the influence of tumors on protective anti-tumor immunity and considers the potential influence that radiation-induced effects might have on the prevalence, phenotype, and function of innate and adaptive immune cells in patients with cancer.
Collapse
Affiliation(s)
- Gemma A Foulds
- Department of Oncology, The Medical School, The University of Sheffield Sheffield, UK ; Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München Munich, Germany
| | | | | | | | | |
Collapse
|
37
|
Liu SL, He W, Wang RP, Li Z, Wu J, Zhao Y, Hu XH. Effects of recombinant hIL-2-Luffin P1 immunotoxin combining with arotinoid ethylester on the proliferation and apoptosis of Hut-78 cells. Mol Cell Toxicol 2013. [DOI: 10.1007/s13273-012-0041-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
A different immunologic profile characterizes patients with HER-2-overexpressing and HER-2-negative locally advanced breast cancer: implications for immune-based therapies. Breast Cancer Res 2012; 13:R117. [PMID: 22112244 PMCID: PMC3326559 DOI: 10.1186/bcr3060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/16/2011] [Accepted: 11/23/2011] [Indexed: 02/01/2023] Open
Abstract
Introduction The clinical efficacy of trastuzumab and taxanes is at least partly related to their ability to mediate or promote antitumor immune responses. On these grounds, a careful analysis of basal immune profile may be capital to dissect the heterogeneity of clinical responses to these drugs in patients with locally advanced breast cancer undergoing neoadjuvant chemotherapy. Methods Blood samples were collected from 61 locally advanced breast cancers (36 HER2- and 25 HER2+) at diagnosis and from 23 healthy women. Immunophenotypic profiling of circulating and intratumor immune cells, including regulatory T (Treg) cells, was assessed by flow cytometry and immunohistochemistry, respectively. Serum levels of 10 different cytokines were assessed by multiplex immunoassays. CD8+ T cell responses to multiple tumor-associated antigens (TAA) were evaluated by IFN-γ-enzyme-linked immunosorbent spot (ELISPOT). The Student's t test for two tailed distributions and the Wilcoxon two-sample test were used for the statistical analysis of the data. Results The proportion of circulating immune effectors was similar in HER2+ patients and healthy donors, whereas higher percentages of natural killer and Treg cells and a lower CD4+/CD8+ T cell ratio (with a prevalence of naïve and central memory CD8+ T cells) were observed in HER2- cases. Higher numbers of circulating CD8+ T cells specific for several HLA-A*0201-restricted TAA-derived peptides were observed in HER2+ cases, together with a higher prevalence of intratumor CD8+ T cells. Serum cytokine profile of HER2+ patients was similar to that of controls, whereas HER2- cases showed significantly lower cytokine amounts compared to healthy women (IL-2, IL-8, IL-6) and HER2+ cases (IL-2, IL-1β, IL-8, IL-6, IL-10). Conclusions Compared to HER2- cases, patients with HER2-overexpressing locally advanced breast cancer show a more limited tumor-related immune suppression. This may account for the clinical benefit achieved in this subset of patients with the use of drugs acting through, but also promoting, immune-mediated effects.
Collapse
|
39
|
Yamada Y, Aoyama A, Tocco G, Boskovic S, Nadazdin O, Alessandrini A, Madsen JC, Cosimi AB, Benichou G, Kawai T. Differential effects of denileukin diftitox IL-2 immunotoxin on NK and regulatory T cells in nonhuman primates. THE JOURNAL OF IMMUNOLOGY 2012; 188:6063-70. [PMID: 22586034 DOI: 10.4049/jimmunol.1200656] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Denileukin diftitox (DD), a fusion protein comprising IL-2 and diphtheria toxin, was initially expected to enhance antitumor immunity by selectively eliminating regulatory T cells (Tregs) displaying the high-affinity IL-2R (α-β-γ trimers). Although DD was shown to deplete some Tregs in primates, its effects on NK cells (CD16(+)CD8(+)NKG2A(+)CD3(-)), which constitutively express the intermediate-affinity IL-2R (β-γ dimers) and play a critical role in antitumor immunity, are still unknown. To address this question, cynomolgus monkeys were injected i.v. with two doses of DD (8 or 18 μg/kg). This treatment resulted in a rapid, but short-term, reduction in detectable peripheral blood resting Tregs (CD4(+)CD45RA(+)Foxp3(+)) and a transient increase in the number of activated Tregs (CD4(+)CD45RA(-)Foxp3(high)), followed by their partial depletion (50-60%). In contrast, all NK cells were deleted immediately and durably after DD administration. This difference was not due to a higher binding or internalization of DD by NK cells compared with Tregs. Coadministration of DD with IL-15, which binds to IL-2Rβ-γ, abrogated DD-induced NK cell deletion in vitro and in vivo, whereas it did not affect Treg elimination. Taken together, these results show that DD exerts a potent cytotoxic effect on NK cells, a phenomenon that might impair its antitumoral properties. However, coadministration of IL-15 with DD could alleviate this problem by selectively protecting potentially oncolytic NK cells, while allowing the depletion of immunosuppressive Tregs in cancer patients.
Collapse
Affiliation(s)
- Yohei Yamada
- Department of Surgery, Transplant Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ghosh AK, Basu S. Tumor macrophages as a target for Capsaicin mediated immunotherapy. Cancer Lett 2012; 324:91-7. [PMID: 22579786 DOI: 10.1016/j.canlet.2012.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 04/26/2012] [Accepted: 05/02/2012] [Indexed: 01/24/2023]
Abstract
Tumor microenvironment contributes to a large extent for failure of immunological destruction of antigenic tumors. Most solid tumors adapt to the microenvironment and escape the host immune system. The dramatic and systemic effectiveness of neuro-immune ligand Capsaicin (CP) in regression of established solid tumors led us to investigate its immunomodulatory role in tumor microenvironment. In this report we demonstrate that CP induced tumor cell apoptosis leads to increased sensitization of the surrounding stroma manifested by enhanced antigen presentation by stromal macrophages and its destruction by tumor specific T-cells. Further, CP injection alters the tumor microenvironment with regards to tumor-infiltrating Treg cells as well as the cytokine milieu at the tumor site. Our data collectively demonstrates that injection of CP sets in motion, a cascade of several independent innate and adaptive immunological events initiated at the tumor environment.
Collapse
Affiliation(s)
- Amiya K Ghosh
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, MC1601, Farmington, CT 06030-1601, United States
| | | |
Collapse
|
41
|
CHOI YOOJIN, YANG KWANGMO, KIM SUNGDAE, YOO YOUNGHYUN, LEE SANGWHA, SEO SUYEONG, SUH HONGSUK, YEE SUNGTAE, JEONG MINHO, JO WOLSOON. Resveratrol analogue HS-1793 induces the modulation of tumor-derived T cells. Exp Ther Med 2012; 3:592-598. [PMID: 22969934 PMCID: PMC3438654 DOI: 10.3892/etm.2012.472] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/20/2012] [Indexed: 12/18/2022] Open
Abstract
Recent advances in the understanding of the mechanisms responsible for tumor progression suggest the possibility to control cancer growth, not only through chemotherapy-induced cancer cell destruction, but also by stimulating anticancer immunity. However, immune tolerance against tumor antigens disturbs diverse forms of immunotherapy. One of the most potent and well-studied tumor-induced immunosuppressive phenotypes found in the tumor microenvironment is the regulatory subpopulation cells (CD4(+)CD25(+)FoxP3(+) Treg cells). Among the great number of natural agents derived from plants and potentially useful for application in the complementary therapy of cancer, resveratrol is gaining attention for its immunomodulating properties in breast cancer, since the ineffectiveness of numerous immunotherapy strategies may be related, in part, to their negative effects on Treg cells. The present study was undertaken to examine whether HS-1793, a synthetic resveratrol analogue free from the restriction of the metabolic instability and high dose requirement of resveratrol, shows a direct effect on immune responses by enhancing lymphocyte proliferation or an immunomodulatory effect by inducing changes in the Treg cell population in FM3A breast tumor-bearing mice. Although HS-1793 had no direct immunostimulatory effect, it dose-dependently decreased IL-2 secretion and increased IL-4 secretion of concanavalin A-stimulated lymphocytes from tumor-bearing mice, which suggest that HS-1793 may induce changes in the subpopulations of tumor-derived T lymphocytes. The CD4(+)CD25(+) cell population from tumor-bearing mice decreased after HS-1793 treatment in a dose-dependent manner, while the CD4(+) T cell population remained unchanged. FoxP3(+)-expressing cells among the CD4(+)CD25(+) population showed a similar pattern. In contrast, the CD8(+) T cell population as well as the interferon (IFN)-γ-expressing CD8(+) T cell population and IFN-γ secretion of splenocytes from tumor-bearing mice were significantly upregulated by HS-1793 treatment. These results suggest that HS-1793 induces the modulation of tumor-derived T lymphocytes, particulary having a suppressive effect on the Treg cell population, likely contributing to enhanced tumor-specific cytotoxic T lymphocyte responses and CD4(+) T cells involving antitumor immunity. Therefore, HS-1793 may serve as a promising adjuvant therapeutic reagent in breast cancer immunotherapy.
Collapse
Affiliation(s)
- YOO JIN CHOI
- Research Center, Dong Nam Institute of Radiological and Medical Sciences, Busan 619-953
| | - KWANG MO YANG
- Research Center, Dong Nam Institute of Radiological and Medical Sciences, Busan 619-953
| | - SUNG DAE KIM
- Research Center, Dong Nam Institute of Radiological and Medical Sciences, Busan 619-953
| | - YOUNG HYUN YOO
- Anatomy and Cell Biology, Dong-A University College of Medicine and MitochondriaHub Regulation Center, Busan 602-714
| | | | | | - HONGSUK SUH
- Department of Chemistry and Chemistry Institute for Functional Materials,Pusan National University, Busan 609-735
| | - SUNG TAE YEE
- Department of Biology, Sunchon National University, Sunchon,
Republic of Korea
| | | | - WOL SOON JO
- Research Center, Dong Nam Institute of Radiological and Medical Sciences, Busan 619-953
| |
Collapse
|
42
|
Gritzapis AD, Voutsas IF, Baxevanis CN. Ontak reduces the immunosuppressive tumor environment and enhances successful therapeutic vaccination in HER-2/neu-tolerant mice. Cancer Immunol Immunother 2012; 61:397-407. [PMID: 21928125 PMCID: PMC11029548 DOI: 10.1007/s00262-011-1113-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 09/04/2011] [Indexed: 01/06/2023]
Abstract
Disrupting tumor-mediated mechanisms suppressing host immunity represents a novel approach to tumor immunotherapy. Depletion of regulatory T cells (Tregs) increases endogenous anti-tumor immunity and the efficacy of active immunotherapy in experimental tumor models. HLA-A2.1/HLA-DR1 (A2.1/DR1) × BALB- neuT+ (neuT+) triple transgenic mice represent an improvement over neuT+ mice for evaluating vaccination regimens to overcome tolerance against HER-2/neu. We questioned whether depletion of Tregs with Denileukin diftitox (Ontak) enhances the efficacy of a therapeutic vaccine consisting of HER-2(85-94) (p85) CTL and HER-2(776-790) (p776) Th peptides against the growth of TUBO.A2 transplantable tumor in male A2.1/DR1 × neuT+ Tg mice. While the therapeutic vaccine primed the tumor-reactive CD8+ CTLs and CD4+ effector T lymphocytes (Teffs) compartment, inducing activation, tumor infiltration, and tumor rejection or delay in tumor growth, treatment with Ontak 1 day prior to vaccination resulted in enhanced CD4+ and CD8+ T-cell-mediated vaccine-specific immune responses in the periphery. This was closely associated with greater infiltration and a striking change in the intratumor balance of Tregs and vaccine-specific CTLs/Teffs that directly correlated with markedly enhanced antitumor activity. The data suggest that Tregs control both CD4+ and CD8+ T-cell activity within the tumor, emphasize the importance of the intratumor ratio of vaccine-specific lymphocytes to Tregs, and demonstrate significant inversion of this ratio and correlation with tumor rejection during Ontak/vaccine immunotherapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Diphtheria Toxin/immunology
- Diphtheria Toxin/pharmacology
- Female
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- HLA-DR1 Antigen/genetics
- HLA-DR1 Antigen/immunology
- Humans
- Immune Tolerance/drug effects
- Immune Tolerance/immunology
- Immunosuppressive Agents/immunology
- Immunosuppressive Agents/pharmacology
- Interleukin-2/immunology
- Interleukin-2/pharmacology
- Male
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Rats
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Tumor Burden/drug effects
- Tumor Burden/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Vaccination/methods
Collapse
Affiliation(s)
- Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, St Savas Cancer Hospital, 171 Alexandras Avenue, 115 22 Athens, Greece
| | - Ioannis F. Voutsas
- Cancer Immunology and Immunotherapy Center, St Savas Cancer Hospital, 171 Alexandras Avenue, 115 22 Athens, Greece
| | - Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, St Savas Cancer Hospital, 171 Alexandras Avenue, 115 22 Athens, Greece
| |
Collapse
|
43
|
Place AE, Jin Huh S, Polyak K. The microenvironment in breast cancer progression: biology and implications for treatment. Breast Cancer Res 2011; 13:227. [PMID: 22078026 PMCID: PMC3326543 DOI: 10.1186/bcr2912] [Citation(s) in RCA: 278] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Breast cancer comprises a heterogeneous group of malignancies derived from the ductal epithelium. The microenvironment of these cancers is now recognized as a critical participant in tumor progression and therapeutic responses. Recent data demonstrate significant gene expression and epigenetic alterations in cells composing the microenvironment during disease progression, which can be explored as biomarkers and targets for therapy. Indeed, gene expression signatures derived from tumor stroma have been linked to clinical outcomes. There is increasing interest in translating our current understanding of the tumor microenvironment to the development of novel therapies.
Collapse
Affiliation(s)
- Andrew E Place
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, D740C, Boston, MA 02215, USA
| | | | | |
Collapse
|
44
|
Depleting regulatory T cells with arginine-rich, cell-penetrating, peptide-conjugated morpholino oligomer targeting FOXP3 inhibits regulatory T-cell function. Cancer Gene Ther 2011; 19:30-7. [PMID: 21997230 DOI: 10.1038/cgt.2011.63] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD4+CD25+regulatory T cells (T(reg)) impair anti-tumor and anti-viral immunity. As there are higher T(reg) levels in cancer patients compared with healthy individuals, there is considerable interest in eliminating them or altering their function as part of cancer or viral immunotherapy strategies. The scurfin transcriptional regulator encoded by the member of the forkhead winged helix protein family (FOXP3) is critical for maintaining the functions of T(reg). We hypothesized that targeting FOXP3 expression with a novel arginine-rich, cell-penetrating, peptide-conjugated phosphorodiamidate morpholino (PPMO) based antisense would eliminate T(reg) and enhance the induction of effector T-cell responses. We observed that the PPMO was taken up by activated T cells in vitro and could downregulate FOXP3 expression, which otherwise increases during antigen-specific T-cell activation. Generation of antigen-specific T cells in response to peptide stimulation was enhanced by pre-treatment of peripheral blood mononuclear cells with the FOXP3-targeted PPMO. In summary, modulation of T(reg) levels using the FOXP3 PPMO antisense-based genomic strategy has the potential to optimize immunotherapy strategies in cancer and viral immunotherapy.
Collapse
|
45
|
Divanovic S, Trompette A, Ashworth JI, Rao MB, Karp CL. Therapeutic enhancement of protective immunity during experimental leishmaniasis. PLoS Negl Trop Dis 2011; 5:e1316. [PMID: 21909452 PMCID: PMC3167777 DOI: 10.1371/journal.pntd.0001316] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 08/01/2011] [Indexed: 12/15/2022] Open
Abstract
Background Leishmaniasis remains a significant cause of morbidity and mortality in the tropics. Available therapies are problematic due to toxicity, treatment duration and emerging drug resistance. Mouse models of leishmaniasis have demonstrated that disease outcome depends critically on the balance between effector and regulatory CD4+ T cell responses, something mirrored in descriptive studies of human disease. Recombinant IL-2/diphtheria toxin fusion protein (rIL-2/DTx), a drug that is FDA-approved for the treatment of cutaneous T cell lymphoma, has been reported to deplete regulatory CD4+ T cells. Methodology/Principal Findings We investigated the potential efficacy of rIL-2/DTx as adjunctive therapy for experimental infection with Leishmania major. Treatment with rIL-2/DTx suppressed lesional regulatory T cell numbers and was associated with significantly increased antigen-specific IFN-γ production, enhanced lesion resolution and decreased parasite burden. Combined administration of rIL-2/DTx and sodium stibogluconate had additive biological and therapeutic effects, allowing for reduced duration or dose of sodium stibogluconate therapy. Conclusions/Significance These data suggest that pharmacological suppression of immune counterregulation using a commercially available drug originally developed for cancer therapy may have practical therapeutic utility in leishmaniasis. Rational reinvestigation of the efficacy of drugs approved for other indications in experimental models of neglected tropical diseases has promise in providing new candidates to the drug discovery pipeline. Leishmaniasis is an infectious disease that causes a large burden of morbidity and mortality in the tropics. Caused by protozoan parasites of the genus Leishmania that are transmitted by sandflies, leishmaniasis causes a wide spectrum of human disease. The severe end of the spectrum, visceral leishmaniasis, causes an annual mortality of approximately 50,000, largely in India and Sudan. Available therapies for leishmaniasis are problematic due to emerging drug resistance, toxicity and/or the need for lengthy courses of treatment. There is thus an urgent need for novel therapeutic approaches to this neglected tropical disease. To address this problem, the authors examined whether a commercially available drug developed for cancer therapy (Ontak), reported to have immunological activity of relevance to the immunobiology of Leishmania infection, exhibited efficacy in mouse models of leishmaniasis. The study found therapeutic efficacy for the drug alone in these models, as well as additive therapeutic efficacy in combination with standard antimicrobial therapy. Rational reinvestigation of the efficacy of already approved drugs in experimental models of neglected tropical diseases has promise in providing needed new candidates to the drug discovery pipeline.
Collapse
Affiliation(s)
- Senad Divanovic
- Division of Molecular Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Aurelien Trompette
- Division of Molecular Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jamie I. Ashworth
- Division of Molecular Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Marepalli B. Rao
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Christopher L. Karp
- Division of Molecular Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
46
|
Persson J, Beyer I, Yumul R, Li Z, Kiem HP, Roffler S, Lieber A. Immuno-therapy with anti-CTLA4 antibodies in tolerized and non-tolerized mouse tumor models. PLoS One 2011; 6:e22303. [PMID: 21779410 PMCID: PMC3136517 DOI: 10.1371/journal.pone.0022303] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 06/23/2011] [Indexed: 11/23/2022] Open
Abstract
Monoclonal antibodies specific for cytotoxic T lymphocyte-associated antigen 4 (anti-CTLA4) are a novel form of cancer immunotherapy. While preclinical studies in mouse tumor models have shown anti-tumor efficacy of anti-CTLA4 injection or expression, anti-CTLA4 treatment in patients with advanced cancers had disappointing therapeutic benefit. These discrepancies have to be addressed in more adequate pre-clinical models. We employed two tumor models. The first model is based on C57Bl/6 mice and syngeneic TC-1 tumors expressing HPV16 E6/E7. In this model, the HPV antigens are neo-antigens, against which no central tolerance exists. The second model involves mice transgenic for the proto-oncogen neu and syngeneic mouse mammary carcinoma (MMC) cells. In this model tolerance to Neu involves both central and peripheral mechanisms. Anti-CTLA4 delivery as a protein or expression from gene-modified tumor cells were therapeutically efficacious in the non-tolerized TC-1 tumor model, but had no effect in the MMC-model. We also used the two tumor models to test an immuno-gene therapy approach for anti-CTLA4. Recently, we used an approach based on hematopoietic stem cells (HSC) to deliver the relaxin gene to tumors and showed that this approach facilitates pre-existing anti-tumor T-cells to control tumor growth in the MMC tumor model. However, unexpectedly, when used for anti-CTLA4 gene delivery in this study, the HSC-based approach was therapeutically detrimental in both the TC-1 and MMC models. Anti-CTLA4 expression in these models resulted in an increase in the number of intratumoral CD1d+ NKT cells and in the expression of TGF-β1. At the same time, levels of pro-inflammatory cytokines and chemokines, which potentially can support anti-tumor T-cell responses, were lower in tumors of mice that received anti-CTLA4-HSC therapy. The differences in outcomes between the tolerized and non-tolerized models also provide a potential explanation for the low efficacy of CTLA4 blockage approaches in cancer immunotherapy trials.
Collapse
Affiliation(s)
- Jonas Persson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ines Beyer
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Roma Yumul
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - ZongYi Li
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Steve Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - André Lieber
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Pathology University of Washington, Washington, United States of America
- * E-mail:
| |
Collapse
|
47
|
Erskine CL, Krco CJ, Hedin KE, Borson ND, Kalli KR, Behrens MD, Heman-Ackah SM, von Hofe E, Wettstein PJ, Mohamadzadeh M, Knutson KL. MHC class II epitope nesting modulates dendritic cell function and improves generation of antigen-specific CD4 helper T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:316-24. [PMID: 21613617 DOI: 10.4049/jimmunol.1100658] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CD4 Th cells are critical to the development of coordinated immune responses to infections and tumors. Th cells are activated through interactions of the TCR with MHC class II complexed with peptide. T cell activation is dependent on the density of MHC peptide complexes as well as the duration of interaction of the TCR with APCs. In this study, we sought to determine whether MHC class II peptides could be modified with amino acid sequences that facilitated uptake and presentation with the goal of improving Th cell activation in vitro and in vivo. A model epitope derived from the murine folate receptor α, a self- and tumor Ag, was modified at its carboxyl terminus with the invariant chain-derived Ii-Key peptide and at its N terminus with a peptide that enhances uptake of Ag by APC. Modification of a peptide resulted in enhanced generation of high-avidity murine folate receptor α T cells that persisted in vivo and homed to sites of Ag deposition. The nesting approach was epitope and species independent and specifically excluded expansion of CD4 regulatory T cells. The resulting Th cells were therapeutic, enhanced in vivo helper activity and had an increased ability to resist tolerizing immune microenvironments. In addition to improved immunoadjuvants, this epitope modification strategy may be useful for enhancing ex vivo and in vivo generation of Th cells for preventing and treating diseases.
Collapse
Affiliation(s)
- Courtney L Erskine
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Krempski J, Karyampudi L, Behrens MD, Erskine CL, Hartmann L, Dong H, Goode EL, Kalli KR, Knutson KL. Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer. THE JOURNAL OF IMMUNOLOGY 2011; 186:6905-13. [PMID: 21551365 DOI: 10.4049/jimmunol.1100274] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Within the ovarian cancer microenvironment, there are several mechanisms that suppress the actions of antitumor immune effectors. Delineating the complex immune microenvironment is an important goal toward developing effective immune-based therapies. A dominant pathway of immune suppression in ovarian cancer involves tumor-associated and dendritic cell (DC)-associated B7-H1. The interaction of B7-H1 with PD-1 on tumor-infiltrating T cells is a widely cited theory of immune suppression involving B7-H1 in ovarian cancer. Recent studies suggest that the B7-H1 ligand, programmed death receptor-1 (PD-1), is also expressed on myeloid cells, complicating interpretations of how B7-H1 regulates DC function in the tumor. In this study, we found that ovarian cancer-infiltrating DCs progressively expressed increased levels of PD-1 over time in addition to B7-H1. These dual-positive PD-1(+) B7-H1(+) DCs have a classical DC phenotype (i.e., CD11c(+)CD11b(+)CD8(-)), but are immature, suppressive, and respond poorly to danger signals. Accumulation of PD-1(+)B7-H1(+) DCs in the tumor was associated with suppression of T cell activity and decreased infiltrating T cells in advancing tumors. T cell suppressor function of these DCs appeared to be mediated by T cell-associated PD-1. In contrast, ligation of PD-1 expressed on the tumor-associated DCs suppressed NF-κB activation, release of immune regulatory cytokines, and upregulation of costimulatory molecules. PD-1 blockade in mice bearing ovarian cancer substantially reduced tumor burden and increased effector Ag-specific T cell responses. Our results reveal a novel role of tumor infiltrating PD-1(+)B7-H1(+) DCs in mediating immune suppression in ovarian cancer.
Collapse
Affiliation(s)
- James Krempski
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Salagianni M, Lekka E, Moustaki A, Iliopoulou EG, Baxevanis CN, Papamichail M, Perez SA. NK Cell Adoptive Transfer Combined with Ontak-Mediated Regulatory T Cell Elimination Induces Effective Adaptive Antitumor Immune Responses. THE JOURNAL OF IMMUNOLOGY 2011; 186:3327-35. [DOI: 10.4049/jimmunol.1000652] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
50
|
Chan T, Wiltrout RH, Weiss JM. Immunotherapeutic modulation of the suppressive liver and tumor microenvironments. Int Immunopharmacol 2011; 11:879-89. [PMID: 21241810 DOI: 10.1016/j.intimp.2010.12.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 12/27/2010] [Indexed: 12/24/2022]
Abstract
The liver is an immunologically unique organ, consisting of resident hematopoietic and parenchymal cells which often contribute to a relatively tolerant microenvironment. It is also becoming increasingly clear that tumor-induced immunosuppression occurs via many of the same cellular mechanisms which contribute to the tolerogenic liver microenvironment. Myeloid cells, consisting of dendritic cells (DC), macrophages and myeloid derived suppressor cells (MDSC), have been implicated in providing a tolerogenic liver environment and immune dysfunction within the tumor microenvironment which can favor tumor progression. As we increase our understanding of the biological mechanisms involved for each phenotypic and/or functionally distinct leukocyte subset, immunotherapeutic strategies can be developed to overcome the inherent barriers to the development of improved strategies for the treatment of liver disease and tumors. In this review, we discuss the principal myeloid cell-based contributions to immunosuppression that are shared between the liver and tumor microenvironments. We further highlight immune-based strategies shown to modulate immunoregulatory cells within each microenvironment and enhance anti-tumor responses.
Collapse
Affiliation(s)
- Tim Chan
- NCI Frederick, Building 560, Room 31-18 Frederick, MD 21702, USA
| | | | | |
Collapse
|