1
|
Dwivedi M, Tiwari S, Kemp EH, Begum R. Implications of regulatory T cells in anti-cancer immunity: from pathogenesis to therapeutics. Heliyon 2022; 8:e10450. [PMID: 36082331 PMCID: PMC9445387 DOI: 10.1016/j.heliyon.2022.e10450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/08/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Regulatory T cells (Tregs) play an essential role in maintaining immune tolerance and suppressing inflammation. However, Tregs present major hurdle in eliciting potent anti-cancer immune responses. Therefore, curbing the activity of Tregs represents a novel and efficient way towards successful immunotherapy of cancer. Moreover, there is an emerging interest in harnessing Treg-based strategies for augmenting anti-cancer immunity in different types of the disease. This review summarises the crucial mechanisms of Tregs’ mediated suppression of anti-cancer immunity and strategies to suppress or to alter such Tregs to improve the immune response against tumors. Highlighting important clinical studies, the review also describes current Treg-based therapeutic interventions in cancer, and discusses Treg-suppression by molecular targeting, which may emerge as an effective cancer immunotherapy and as an alternative to detrimental chemotherapeutic agents. Tregs are crucial in maintaining immune tolerance and suppressing inflammation. Tregs present a major obstacle to eliciting potent anti-tumor immune responses. The review summarizes current Treg-based therapeutic interventions in cancer. Treg can be an effective cancer immunotherapy target.
Collapse
Affiliation(s)
- Mitesh Dwivedi
- C. G. Bhakta Institute of Biotechnology, Faculty of Science, Uka Tarsadia University, Tarsadi, Surat, Gujarat, 394350, India
- Corresponding author.
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, 226002, Uttar Pradesh, India
| | - E. Helen Kemp
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, S10 2RX, UK
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
| |
Collapse
|
2
|
Laplagne C, Domagala M, Le Naour A, Quemerais C, Hamel D, Fournié JJ, Couderc B, Bousquet C, Ferrand A, Poupot M. Latest Advances in Targeting the Tumor Microenvironment for Tumor Suppression. Int J Mol Sci 2019; 20:E4719. [PMID: 31547627 PMCID: PMC6801830 DOI: 10.3390/ijms20194719] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
The tumor bulk is composed of a highly heterogeneous population of cancer cells, as well as a large variety of resident and infiltrating host cells, extracellular matrix proteins, and secreted proteins, collectively known as the tumor microenvironment (TME). The TME is essential for driving tumor development by promoting cancer cell survival, migration, metastasis, chemoresistance, and the ability to evade the immune system responses. Therapeutically targeting tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), regulatory T-cells (T-regs), and mesenchymal stromal/stem cells (MSCs) is likely to have an impact in cancer treatment. In this review, we focus on describing the normal physiological functions of each of these cell types and their behavior in the cancer setting. Relying on the specific surface markers and secreted molecules in this context, we review the potential targeting of these cells inducing their depletion, reprogramming, or differentiation, or inhibiting their pro-tumor functions or recruitment. Different approaches were developed for this targeting, namely, immunotherapies, vaccines, small interfering RNA, or small molecules.
Collapse
Affiliation(s)
- Chloé Laplagne
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Marcin Domagala
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Augustin Le Naour
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France.
| | - Christophe Quemerais
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Dimitri Hamel
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut de Recherche en Santé Digestive, Inserm U1220, INRA, ENVT, 31024 Toulouse, France.
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Bettina Couderc
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France.
| | - Corinne Bousquet
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Audrey Ferrand
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut de Recherche en Santé Digestive, Inserm U1220, INRA, ENVT, 31024 Toulouse, France.
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| |
Collapse
|
3
|
Osella-Abate S, Conti L, Annaratone L, Senetta R, Bertero L, Licciardello M, Caliendo V, Picciotto F, Quaglino P, Cassoni P, Ribero S. Phenotypic characterisation of immune cells associated with histological regression in cutaneous melanoma. Pathology 2019; 51:487-493. [PMID: 31266597 DOI: 10.1016/j.pathol.2019.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/18/2019] [Accepted: 03/31/2019] [Indexed: 10/26/2022]
Abstract
Histological regression and tumour infiltrating lymphocytes represent an early sign of activation of the immune system against primary melanoma. The first phenomenon has been especially discussed in the literature because of its prognostic role, but no clear agreement on its evaluation has been reached. Immunotherapy of advanced stage melanoma has recently shown promising results; an improved understanding of the initial interplay between melanoma cells and the immune system would potentially help tailor treatment for patients. Seventy consecutive melanomas with regression were analysed to identify a prognostic cut-off value of regression extension. Then, we compared the immune infiltrate between regressed and not regressed areas of these regressed melanomas, assessing CD3, CD4, CD8, CD20, CD123, PD1 and FOXP3/CD25 expression. The immune infiltrate of these cases was further compared with 28 control melanomas without regression. A regression extension of 10% represented a reliable cut-off to distinguish two different risk categories in regressed melanomas. Regressed areas were less infiltrated by CD4/CD25, FOXP3/CD4 or PD1/CD4 compared to not regressed areas of each sample. These lymphocyte subsets are associated with anergy and hamper the immune CD8+ response towards the cancer cells. Moreover, the relevance of these findings was further supported by the observation that not regressed controls were significantly more infiltrated by these anergic immune cell subsets compared to the regressed cases. These results help understand the real meaning of regression in melanoma. Moreover, the association here identified between specific immunomodulatory immune cell subsets and regression could help in developing new therapeutic strategies.
Collapse
Affiliation(s)
- Simona Osella-Abate
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy
| | - Luca Conti
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy
| | - Laura Annaratone
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy
| | - Rebecca Senetta
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy
| | - Luca Bertero
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy
| | - Matteo Licciardello
- Department of Medical Sciences, Section of Dermatology, University of Turin, Torino, Italy
| | - Virginia Caliendo
- Dermatologic Surgery Section, Department of Oncology, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Franco Picciotto
- Dermatologic Surgery Section, Department of Oncology, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Pietro Quaglino
- Department of Medical Sciences, Section of Dermatology, University of Turin, Torino, Italy
| | - Paola Cassoni
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Torino, Italy.
| | - Simone Ribero
- Department of Medical Sciences, Section of Dermatology, University of Turin, Torino, Italy; Dermatologic Surgery Section, Department of Oncology, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
4
|
Mousavi-Niri N, Naseroleslami M, Hadjati J. Anti-regulatory T cell vaccines in immunotherapy: focusing on FoxP3 as target. Hum Vaccin Immunother 2019; 15:620-624. [PMID: 30633616 PMCID: PMC6605713 DOI: 10.1080/21645515.2018.1545625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/29/2022] Open
Abstract
Anti- tumor vaccination elicits imperfect immune responses against tumor cells; that is related to the presence of suppressive obstacles in the tumor microenvironment. The main members of suppressive milieu of tumor are heteroogenous groups of immune cells in which regulatory T cell is a substantial component. Tregs express different immunomodulatory molecules such as FoxP3. Transcription factor, FoxP3, is a specific intracellular marker of Treg and crucial for Treg development. Therefore it is an attractive target for cancer treatment. This article reviews some recent anti-Treg vaccine focusing on FoxP3 to ameliorate anti-tumor immune responses. Among them, fusion vaccine of FoxP3-Fc(IgG) recombinant DNA vaccine and its accordant protein vaccine represents effective results.
Collapse
Affiliation(s)
- Neda Mousavi-Niri
- Department of Medical Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Naseroleslami
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Guerrero S, Inostroza-Riquelme M, Contreras-Orellana P, Diaz-Garcia V, Lara P, Vivanco-Palma A, Cárdenas A, Miranda V, Robert P, Leyton L, Kogan MJ, Quest AFG, Oyarzun-Ampuero F. Curcumin-loaded nanoemulsion: a new safe and effective formulation to prevent tumor reincidence and metastasis. NANOSCALE 2018; 10:22612-22622. [PMID: 30484463 DOI: 10.1039/c8nr06173d] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Curcumin is widely considered beneficial to human health, but insolubility and instability greatly hamper reproducible exploitation of the advantageous traits. Here we report on the development, characterization and evaluation of a curcumin-loaded nanoemulsion (CUR-NEM) that is highly effective in preventing post-surgery tumor reincidence and metastasis. The method of fabrication utilized safe excipients and generated particles of 200 nm (PDI ≤ 0.2) with negative zeta potential (-30 mV) and a high yield of curcumin (95%), which can be converted by lyophilization to a dry powder. In vitro assays showed that CUR-NEM is safe in non-cancerous human cells (HEK-293T) and preferentially cytotoxic in gastric (AGS), colon (HT29-ATCC, HT29-US), breast (MDA-MB-231) and melanoma (B16F10) cells. In addition, in melanoma cells the nanoformulation increases intracellular curcumin accumulation and reactive oxygen species (ROS) formation, while preventing cell-migration and invasion. In vivo studies in C57BL/6 mice demonstrated that a single dose, applied topically to the wounded area after surgical excision of primary tumors formed upon subcutaneous injection of syngeneic B16F10 cells, was sufficient to completely prevent reincident tumor growth and spontaneous lung metastasis, while in untreated animals 70% reincidence and metastasis were observed. In vivo experiments also showed that the fluorescence signal due to curcumin was maintained at least 15 days after topical application of CUR-NEM, while when administered in DMSO the curcumin signal disappeared within 4 days. Importantly, the administration of a dose 22 times larger than that applied topically to animals after tumor surgery did not alter biochemical parameters. Due to the safety and efficacy of the formulation, we envisage it as ideal for topical application in cancer patients following surgery, to prevent tumor reincidence and metastasis. In addition, other routes of administration/protocols could also be proposed to treat/prevent malignant tumors in patients.
Collapse
Affiliation(s)
- Simón Guerrero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 8380453, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Porcellato I, Brachelente C, De Paolis L, Menchetti L, Silvestri S, Sforna M, Vichi G, Iussich S, Mechelli L. FoxP3 and IDO in Canine Melanocytic Tumors. Vet Pathol 2018; 56:189-199. [DOI: 10.1177/0300985818808530] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human melanoma is one of the deadliest forms of cancer, with poor prognosis and high resistance to chemotherapy and radiotherapy. The discovery of immunosuppressive mechanisms in the human melanoma microenvironment led to the use of new prognostic markers and to the development of immunotherapies targeting immune checkpoint molecules. Immunoescape mechanisms in canine melanoma have not yet been investigated, and no such immunotherapy has been tested. The aim of this study was to provide preliminary data on the expression of transcription factor forkhead box protein P3 (FoxP3) and indoleamine 2,3-dioxygenase (IDO) in primary canine melanocytic tumors and to investigate their prognostic role. Formalin-fixed, paraffin-embedded samples from 74 canine melanocytic tumors (26 oral melanomas, 23 cutaneous melanomas, and 25 cutaneous melanocytomas) were retrospectively evaluated by immunohistochemistry to explore the expression of FoxP3 and IDO. An increased risk of death due to melanoma was associated with a higher number of FoxP3+ cells per high-power field (FoxP3+/HPF), a higher percentage of CD3+ cells that were also FoxP3+ infiltrating and surrounding the tumor (%FoxP3), and a higher number of IDO+ cells/HPF (IDO+/HPF). A prognostic value for FoxP3 and IDO is suggested by our study, with optimal cutoffs of 14.7 FoxP3+ cells/HPF, 6.1 IDO+ cells/HPF, and 12.5% FoxP3+ cells. Both markers were also associated with tumor type. Multivariable analysis identified IDO+/HPF ( P < .001) as an independent prognostic marker. Even though stratification by diagnosis caused a loss of significance, results from the present study suggest a prognostic role for IDO and FoxP3, possibly related to the establishment of an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Chiara Brachelente
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Livia De Paolis
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Laura Menchetti
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | - Monica Sforna
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Gaia Vichi
- Laboratorio per Analisi Veterinarie Cimie, Macerata, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Torino, Grugliasco (TO), Italy
| | - Luca Mechelli
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
7
|
Ocanha-Xavier JP, Xavier-Junior JCC, Marques MEA. Usefulness of IMP3 and FOXP3 to predict metastasis of cutaneous melanomas. SURGICAL AND EXPERIMENTAL PATHOLOGY 2018. [DOI: 10.1186/s42047-018-0014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Melanoma still has considerable mortality in spite of improvements in diagnosis and treatment. Unfortunately, current diagnostic procedures cannot predict precisely its biological behavior, what urges specialists in searching new better biomarkers of lousy prognosis. The objective of the study was to evaluate IMP3 and FOXP3 expression in primary skin melanoma lesions and to correlate with the presence of metastasis.
Methods
A retrospective cohort study analyzed 112 patients diagnosed with Melanoma, from 2003 to 2011, from a public health service. Samples from the primary lesion were analyzed by two pathologists and one dermatologist to ensure histological subtype, Breslow, the presence of ulceration, mitosis and histological regression. From the species stored, FOXP3 and IMP3 immunohistochemistry staining were performed. Demographic, clinical and evolution aspects of the patients were obtained from records, in the year of 2015. It was considered statistically significant when p-value < 0.05.
Results
The majority of specimens had 25% or fewer cells stained with FOXP3 or IMP3. Their positivity could not be related to the occurrence of metastasis (p = 0.947 and p = 0.936, respectively).
Conclusion
There is no evidence of benefit in using IMP3 or FOXP3 as prognostic markers in primary melanomas in our population.
Collapse
|
8
|
Yonekura S, Itoh M, Shiratori E, Ohtaka M, Tohda S. FOXP3 knockdown inhibits the proliferation and reduces NOTCH1 expression of T cell acute lymphoblastic leukemia cells. BMC Res Notes 2018; 11:582. [PMID: 30103821 PMCID: PMC6090594 DOI: 10.1186/s13104-018-3700-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/09/2018] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Forkhead box P3 (FOXP3) is a master transcriptional factor of regulatory T-cells (Tregs). Recent studies have shown that FOXP3 is associated with growth inhibition of cancer cells. However, the role of FOXP3 in acute T-lymphoblastic leukemia (T-ALL) cells is not known. It was also reported that NOTCH signaling promoted the expression of FOXP3 in Tregs. However, the effect of FOXP3 on NOTCH expression in T-ALL cells is little known. Therefore, we examined the effect of FOXP3 knockdown on the proliferation of T-ALL cells and NOTCH1 signaling. RESULTS Two T-ALL cell lines Jurkat and KOPT-K1, harboring activating NOTCH1 mutations, were transfected with small interfering RNA against FOXP3. Cell growth was assessed with a colorimetric assay and morphology was observed under a microscope. FOXP3 knockdown significantly reduced cell growth and induced morphological changes suggesting apoptosis. Quantitative polymerase chain reaction revealed that FOXP3 knockdown caused the downregulation of mRNA expression of NOTCH1 and HES1. These findings suggest that FOXP3 supports the growth of T-ALL cells although this can not be generalized because we examined only two cell lines. The observed growth suppression can be partly due to the downregulation of NOTCH1 signaling. FOXP3 may be a potential therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Satoru Yonekura
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mai Itoh
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Erika Shiratori
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mika Ohtaka
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Shuji Tohda
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
9
|
Skarmoutsou E, Bevelacqua V, D' Amico F, Russo A, Spandidos DA, Scalisi A, Malaponte G, Guarneri C. FOXP3 expression is modulated by TGF‑β1/NOTCH1 pathway in human melanoma. Int J Mol Med 2018; 42:392-404. [PMID: 29620159 PMCID: PMC5979787 DOI: 10.3892/ijmm.2018.3618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Forkhead box protein 3 (FOXP3) transcription factor is expressed by immune cells and several human cancers and is associated with tumor aggressiveness and unfavorable clinical outcomes. NOTCH and transforming growth factor-β (TGF-β) protumorigenic effects are mediated by FOXP3 expression in several cancer models; however, their interaction and role in melanoma is unknown. We investigated TGF-β-induced FOXP3 gene expression during NOTCH1 signaling inactivation. Primary (WM35) and metastatic melanoma (A375 and A2058) cell lines and normal melanocytes (NHEM) were used. FOXP3 subcellular distribution was evaluated by immuno cytochemical analysis. Gene expression levels were assessed by reverse transcription-quantitative polymerase chain reaction. Protein levels were assessed by western blot analysis. The γ-secretase inhibitor (GSI) was used for NOTCH1 inhibition and recombinant human (rh)TGF-β was used for melanoma cell stimulation. Cell proliferation and viability were respectively assessed by MTT and Trypan blue dye assays. FOXP3 mRNA and protein levels were progressively higher in WM35, A375 and A2058 cell lines compared to NHEM and their levels were further increased after stimulation with rh-TGF-β. TGF-β-mediated FOXP3 expression was mediated by NOTCH1 signaling. Inhibition of NOTCH1 with concomitant rh-TGF-β stimulation determined the reduction in gene expression and protein level of FOXP3. Finally, melanoma cell line proliferation and viability were reduced by NOTCH1 inhibition. The results show that nn increase in FOXP3 expression in metastatic melanoma cell lines is a potential marker of tumor aggressiveness and metastasis. NOTCH1 is a central mediator of TGF-β-mediated FOXP3 expression and NOTCH1 inhibition produces a significant reduction of melanoma cell proliferation and viability.
Collapse
Affiliation(s)
- Eva Skarmoutsou
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Valentina Bevelacqua
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Fabio D' Amico
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Angela Russo
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Crete, Greece
| | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP‑Catania, 95100 Catania, Italy
| | - Grazia Malaponte
- Research Unit of the Catania Section of the Italian League Against Cancer, 95122 Catania, Italy
| | - Claudio Guarneri
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98122 Messina, Italy
| |
Collapse
|
10
|
Nuclear galectin-1-FOXP3 interaction dampens the tumor-suppressive properties of FOXP3 in breast cancer. Cell Death Dis 2018; 9:416. [PMID: 29549328 PMCID: PMC5856744 DOI: 10.1038/s41419-018-0448-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/08/2018] [Accepted: 02/19/2018] [Indexed: 12/22/2022]
Abstract
FOXP3 is an important X-linked suppressor of breast cancer. It is reported that FOXP3 is usually mutant, absent, or cytoplasmic distribution in breast cancer cells, which increases the risk of breast cancer. However, in our study the full-length FOXP3 transcript can be detected in breast cancer cells and nuclear FOXP3 is expressed in some breast cancer samples. Therefore, an important question is how the tumor-suppressive function of wild-type FOXP3 is negated in these cancers. We found that Gal-1 is a novel interacting protein of FOXP3 in breast cancer. Furthermore, our results show that the FKH domain in FOXP3 is essential for its interaction with Gal-1. Through ChIP-seq assay, we found that the expression of Gal-1 could inhibit a variety of target genes which were directly regulated by FOXP3. More importantly, these FOXP3-bound genes are involved in the development and metastasis of cancer. Furthermore, functional studies revealed that blocking the FOXP3/Gal-1 interaction restores the tumor-suppressive properties of FOXP3 in breast cancer cells. Finally, we observed that the nuclear abundance of Gal-1 was significantly higher in breast cancer tissues than that in adjacent normal tissues. In addition, we identified that the acidic extracellular microenvironment in breast cancer tissues causes Gal-1 to accumulate in the nucleus. Altogether, nuclear Gal-1 interferes with the binding of FOXP3 to DNA by interacting with the FKH domain of FOXP3, and it indicates a possible mechanism for the loss of the tumor-suppressive properties of FOXP3 in wild-type FOXP3-positive breast cancer.
Collapse
|
11
|
Namdar A, Mirzaei R, Memarnejadian A, Boghosian R, Samadi M, Mirzaei HR, Farajifard H, Zavar M, Azadmanesh K, Elahi S, Noorbakhsh F, Rezaei A, Hadjati J. Prophylactic DNA vaccine targeting Foxp3 + regulatory T cells depletes myeloid-derived suppressor cells and improves anti-melanoma immune responses in a murine model. Cancer Immunol Immunother 2018; 67:367-379. [PMID: 29124314 PMCID: PMC11028379 DOI: 10.1007/s00262-017-2088-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/05/2017] [Indexed: 01/04/2023]
Abstract
Regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) are the two important and interactive immunosuppressive components of the tumor microenvironment that hamper anti-tumor immune responses. Therefore, targeting these two populations together might be beneficial for overcoming immune suppression in the tumor microenvironment. We have recently shown that prophylactic Foxp3 DNA/recombinant protein vaccine (Foxp3 vaccine) promotes immunity against Treg in tumor-free conditions. In the present study, we investigated the immune modulatory effects of a prophylactic regimen of the redesigned Foxp3 vaccine in the B16F10 melanoma model. Our results indicate that Foxp3 vaccination continuously reduces Treg population in both the tumor site and the spleen. Surprisingly, Treg reduction was associated with a significant decrease in the frequency of MDSC, both in the spleen and in the tumor environment. Furthermore, Foxp3 vaccination resulted in a significant reduction of arginase-1(Arg-1)-induced nitric oxide synthase (iNOS), reactive oxygen species (ROS) and suppressed MDSC activity. Moreover, this concurrent depletion restored production of inflammatory cytokine IFN-γ and enhanced tumor-specific CTL response, which subsequently resulted in the reduction of tumor growth and the improved survival rate of vaccinated mice. In conclusion, our results revealed that Foxp3 vaccine promotes an immune response against tumor by targeting both Treg and MDSC, which could be exploited as a potential immunotherapy approach.
Collapse
Affiliation(s)
- Afshin Namdar
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Reza Mirzaei
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
| | | | - Roobina Boghosian
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
| | - Morteza Samadi
- Recurrent Abortion Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamid Reza Mirzaei
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hamid Farajifard
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
| | - Mehdi Zavar
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
| | | | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Farshid Noorbakhsh
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran
| | - Abbas Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jamshid Hadjati
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, 14155-6447, Iran.
| |
Collapse
|
12
|
Moreno Ayala MA, Gottardo MF, Imsen M, Asad AS, Bal de Kier Joffé E, Casares N, Lasarte JJ, Seilicovich A, Candolfi M. Therapeutic blockade of Foxp3 in experimental breast cancer models. Breast Cancer Res Treat 2017; 166:393-405. [PMID: 28756536 DOI: 10.1007/s10549-017-4414-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/22/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE Regulatory T cells (Tregs) impair the clinical benefit of cancer immunotherapy. To optimize the antitumor efficacy of therapeutic dendritic cell (DC) vaccines, we aimed to inhibit Foxp3, a transcription factor required for Treg function. METHODS Mice bearing established syngeneic LM3 and 4T1 breast tumors were treated with antitumor DC vaccines and a synthetic peptide (P60) that has been shown to inhibit Foxp3. RESULTS Treatment with P60 improved the therapeutic efficacy of DC vaccines in these experimental models. In addition, monotherapy with P60 inhibited tumor growth in immunocompetent as well as in immuno-compromised animals bearing established tumors. We found expression of Foxp3 in human and murine breast tumor cells. P60 inhibited IL-10 secretion in breast cancer cells that expressed Foxp3. CONCLUSIONS Our results suggest that Foxp3 blockade improves the therapeutic efficacy of DC vaccines by inhibition of Tregs and through a direct antitumor effect. This strategy could prove useful to neutralize the immunosuppressive microenvironment and to boost antitumor immunity in breast cancer.
Collapse
Affiliation(s)
- Mariela A Moreno Ayala
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina
| | - María Florencia Gottardo
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina.,Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Imsen
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina
| | - Antonela S Asad
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina
| | - Elisa Bal de Kier Joffé
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Área Investigación, Instituto de Oncología Angel H. Roffo, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Noelia Casares
- Program Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), Pamplona, Avenida Pio XII 55, 31008, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
| | - Juan José Lasarte
- Program Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), Pamplona, Avenida Pio XII 55, 31008, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina.,Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, CONICET, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, C1121ABG, Argentina.
| |
Collapse
|
13
|
Miguel A, Sendra L, Noé V, Ciudad CJ, Dasí F, Hervas D, Herrero MJ, Aliño SF. Silencing of Foxp3 enhances the antitumor efficacy of GM-CSF genetically modified tumor cell vaccine against B16 melanoma. Onco Targets Ther 2017; 10:503-514. [PMID: 28176947 PMCID: PMC5271385 DOI: 10.2147/ott.s104393] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The antitumor response after therapeutic vaccination has a limited effect and seems to be related to the presence of T regulatory cells (Treg), which express the immunoregulatory molecules CTLA4 and Foxp3. The blockage of CTLA4 using antibodies has shown an effective antitumor response conducing to the approval of the human anti-CTLA4 antibody ipilimumab by the US Food and Drug Administration. On the other hand, Foxp3 is crucial for Treg development. For this reason, it is an attractive target for cancer treatment. This study aims to evaluate whether combining therapeutic vaccination with CTLA4 or Foxp3 gene silencing enhances the antitumor response. First, the "in vitro" cell entrance and gene silencing efficacy of two tools, 2'-O-methyl phosphorotioate-modified oligonucleotides (2'-OMe-PS-ASOs) and polypurine reverse Hoogsteen hairpins (PPRHs), were evaluated in EL4 cells and cultured primary lymphocytes. Following B16 tumor transplant, C57BL6 mice were vaccinated with irradiated B16 tumor cells engineered to produce granulocyte-macrophage colony-stimulating factor (GM-CSF) and were intraperitoneally treated with CTLA4 and Foxp3 2'-OMe-PS-ASO before and after vaccination. Tumor growth, mice survival, and CTLA4 and Foxp3 expression in blood cells were measured. The following results were obtained: 1) only 2'-OMe-PS-ASO reached gene silencing efficacy "in vitro"; 2) an improved survival effect was achieved combining both therapeutic vaccine and Foxp3 antisense or CTLA4 antisense oligonucleotides (50% and 20%, respectively); 3) The blood CD4+CD25+Foxp3+ (Treg) and CD4+CTLA4+ cell counts were higher in mice that developed tumor on the day of sacrifice. Our data showed that tumor cell vaccine combined with Foxp3 or CTLA4 gene silencing can increase the efficacy of therapeutic antitumor vaccination.
Collapse
Affiliation(s)
- Antonio Miguel
- Department of Pharmacology, Faculty of Medicine, University of Valencia
| | - Luis Sendra
- Department of Pharmacology, Faculty of Medicine, University of Valencia
| | - Verónica Noé
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Barcelona
| | - Carles J Ciudad
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Barcelona
| | - Francisco Dasí
- Research University Hospital of Valencia, INCLIVA Health Research Institute; Department of Physiology, Faculty of Medicine, University of Valencia Foundation
| | | | - María José Herrero
- Department of Pharmacology, Faculty of Medicine, University of Valencia; Pharmacogenetics Unit, Instituto de Investigación Sanitaria La Fe (IIS La Fe)
| | - Salvador F Aliño
- Department of Pharmacology, Faculty of Medicine, University of Valencia; Clinical Pharmacology Unit, ACM Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|