1
|
Zaakouk M, Longworth A, Hunter K, Jiman S, Kearns D, El-Deftar M, Shaaban AM. Detailed Profiling of the Tumor Microenvironment in Ethnic Breast Cancer, Using Tissue Microarrays and Multiplex Immunofluorescence. Int J Mol Sci 2024; 25:6501. [PMID: 38928207 PMCID: PMC11203983 DOI: 10.3390/ijms25126501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer poses a global health challenge, yet the influence of ethnicity on the tumor microenvironment (TME) remains understudied. In this investigation, we examined immune cell infiltration in 230 breast cancer samples, emphasizing diverse ethnic populations. Leveraging tissue microarrays (TMAs) and core samples, we applied multiplex immunofluorescence (mIF) to dissect immune cell subtypes across TME regions. Our analysis revealed distinct immune cell distribution patterns, particularly enriched in aggressive molecular subtypes triple-negative and HER2-positive tumors. We observed significant correlations between immune cell abundance and key clinicopathological parameters, including tumor size, lymph node involvement, and patient overall survival. Notably, immune cell location within different TME regions showed varying correlations with clinicopathologic parameters. Additionally, ethnicities exhibited diverse distributions of cells, with certain ethnicities showing higher abundance compared to others. In TMA samples, patients of Chinese and Caribbean origin displayed significantly lower numbers of B cells, TAMs, and FOXP3-positive cells. These findings highlight the intricate interplay between immune cells and breast cancer progression, with implications for personalized treatment strategies. Moving forward, integrating advanced imaging techniques, and exploring immune cell heterogeneity in diverse ethnic cohorts can uncover novel immune signatures and guide tailored immunotherapeutic interventions, ultimately improving breast cancer management.
Collapse
Affiliation(s)
- Mohamed Zaakouk
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (M.Z.); (K.H.)
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK; (A.L.); (S.J.); (D.K.)
- Cancer Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Aisling Longworth
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK; (A.L.); (S.J.); (D.K.)
| | - Kelly Hunter
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (M.Z.); (K.H.)
| | - Suhaib Jiman
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK; (A.L.); (S.J.); (D.K.)
| | - Daniel Kearns
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK; (A.L.); (S.J.); (D.K.)
| | - Mervat El-Deftar
- Cancer Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Abeer M Shaaban
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; (M.Z.); (K.H.)
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK; (A.L.); (S.J.); (D.K.)
| |
Collapse
|
2
|
Ibrahim YS, Amin AH, Jawhar ZH, Alghamdi MA, Al-Awsi GRL, Shbeer AM, Al-Ghamdi HS, Gabr GA, Ramírez-Coronel AA, Almulla AF. "To be or not to Be": Regulatory T cells in melanoma. Int Immunopharmacol 2023; 118:110093. [PMID: 37023699 DOI: 10.1016/j.intimp.2023.110093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
In spite of progresses in the therapy of different malignancies, melanoma still remains as one of lethal types of skin tumor. Melanoma is almost easily treatable by surgery alone with higher overall survival rates when it is diagnosed at early stages. However, survival rates are decreased remarkably upon survival if the tumor is progressed to advanced metastatic stages. Immunotherapeutics have been prosperous in the development of anti-tumor responses in patients with melanoma through promotion of the tumor-specific effector T cells in vivo; nonetheless, suitable clinical outcomes have not been satisfactory. One of the underlying causes of the unfavorable clinical outcomes might stem from adverse effects of regulatory T (Treg) cell, which is a prominent mechanism of tumor cells to escape from tumor-specific immune responses. Evidence shows that a poor prognosis and low survival rate in patients with melanoma can be attributed to a higher Treg cell number and function in these subjects. As a result, to promote melanoma-specific anti-tumor responses, depletion of Treg cells appears to be a promising approach; even though the clinical efficacy of different approaches to attain appropriate Treg cell depletion has been inconsistent. Here in this review, the main purpose is to assess the role of Treg cells in the initiation and perpetuation of melanoma and to discuss effective strategies for Treg cell modulation with the aim of melanoma therapy.
Collapse
Affiliation(s)
- Yousif Saleh Ibrahim
- Department of Medical Laboratory Techniques, Al-maarif University College, Ramadi, Al-Anbar, Iraq
| | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq; Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Mohammad A Alghamdi
- Internal Medicine Department, Faculty of Medicine, Albaha University, Saudi Arabia
| | | | - Abdullah M Shbeer
- Department of Surgery, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| | - Hasan S Al-Ghamdi
- Internal Medicine Department, Division of Dermatology, Faculty of Medicine, Albaha University, Albaha City, Saudi Arabia
| | - Gamal A Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center, Giza, Egypt.
| | - Andrés Alexis Ramírez-Coronel
- Catholic University of Cuenca, Azogues Campus, Ecuador; University of Palermo, Buenos Aires, Argentina; National University of Education, Azogues, Ecuador; CES University, Colombia
| | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| |
Collapse
|
3
|
Regulatory T Cell Depletion Using a CRISPR Fc-Optimized CD25 Antibody. Int J Mol Sci 2022; 23:ijms23158707. [PMID: 35955841 PMCID: PMC9369266 DOI: 10.3390/ijms23158707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Regulatory T cells (Tregs) are major drivers behind immunosuppressive mechanisms and present a major hurdle for cancer therapy. Tregs are characterized by a high expression of CD25, which is a potentially valuable target for Treg depletion to alleviate immune suppression. The preclinical anti-CD25 (αCD25) antibody, clone PC-61, has met with modest anti-tumor activity due to its capacity to clear Tregs from the circulation and lymph nodes, but not those that reside in the tumor. The optimization of the Fc domain of this antibody clone has been shown to enhance the intratumoral Treg depletion capacity. Here, we generated a stable cell line that produced optimized recombinant Treg-depleting antibodies. A genome engineering strategy in which CRISPR-Cas9 was combined with homology-directed repair (CRISPR-HDR) was utilized to optimize the Fc domain of the hybridoma PC-61 for effector functions by switching it from its original rat IgG1 to a mouse IgG2a isotype. In a syngeneic tumor mouse model, the resulting αCD25-m2a (mouse IgG2a isotype) antibody mediated the effective depletion of tumor-resident Tregs, leading to a high effector T cell (Teff) to Treg ratio. Moreover, a combination of αCD25-m2a and an αPD-L1 treatment augmented tumor eradication in mice, demonstrating the potential for αCD25 as a cancer immunotherapy.
Collapse
|
4
|
Wasiuk A, Weidlick J, Sisson C, Widger J, Crocker A, Vitale L, Marsh HC, Keler T, He LZ. Conditioning treatment with CD27 Ab enhances expansion and antitumor activity of adoptively transferred T cells in mice. Cancer Immunol Immunother 2021; 71:97-109. [PMID: 34028568 PMCID: PMC8739312 DOI: 10.1007/s00262-021-02958-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/03/2021] [Indexed: 11/17/2022]
Abstract
Cyclophosphamide plus fludarabine (C/F) are currently used to improve the expansion and effectiveness of adoptive cell therapy (ACT). However, these chemotherapeutics cause pan-leukopenia and adverse events, suggesting that safer and more effective conditioning treatments are needed to improve ACT outcomes. Previously, we reported that varlilumab, a CD27-targeting antibody, mediates Treg -preferential T cell depletion, CD8-T cell dominant costimulation, and systemic immune activation in hCD27 transgenic mice and cancer patients. We reasoned that the activities induced by varlilumab may provide an effective conditioning regimen for ACT. Varlilumab pretreatment of hCD27+/+mCD27 − /− mice resulted in prominent proliferation of transferred T cells isolated from wild-type mice. These studies uncovered a critical role for CD27 signaling for the expansion of transferred T cells, as transfer of T cells from CD27 deficient mice or treatment with a CD70 blocking antibody greatly reduced their proliferation. In this model, varlilumab depletes endogenous hCD27+/+ T cells and blocks their subsequent access to CD70, allowing for more CD70 costimulation available to the mCD27+/+ transferred T cells. CD27-targeted depletion led to a greater expansion of transferred T cells compared to C/F conditioning and resulted in longer median survival and more cures than C/F conditioning in the E.G7 tumor model receiving OT-I cell therapy. We propose that translation of this work could be achieved through engineering of T cells for ACT to abrogate varlilumab binding but preserve CD70 ligation. Thus, varlilumab could be an option to chemotherapy as a conditioning regimen for ACT.
Collapse
Affiliation(s)
- Anna Wasiuk
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Jeff Weidlick
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Crystal Sisson
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Jenifer Widger
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Andrea Crocker
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Laura Vitale
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Henry C Marsh
- Celldex Therapeutics, Inc., Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - Tibor Keler
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States
| | - Li-Zhen He
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, United States.
| |
Collapse
|
5
|
Adalid-Peralta L, Lopez-Roblero A, Camacho-Vázquez C, Nájera-Ocampo M, Guevara-Salinas A, Ruiz-Monroy N, Melo-Salas M, Morales-Ruiz V, López-Recinos D, Ortiz-Hernández E, Demengeot J, Vazquez-Perez JA, Arce-Sillas A, Gomez-Fuentes S, Parkhouse RME, Fragoso G, Sciutto E, Sevilla-Reyes EE. Regulatory T Cells as an Escape Mechanism to the Immune Response in Taenia crassiceps Infection. Front Cell Infect Microbiol 2021; 11:630583. [PMID: 33928043 PMCID: PMC8076859 DOI: 10.3389/fcimb.2021.630583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/22/2021] [Indexed: 11/18/2022] Open
Abstract
Murine cysticercosis by Taenia crassiceps is a model for human neurocysticercosis. Genetic and/or immune differences may underlie the higher susceptibility to infection in BALB/cAnN with respect to C57BL/6 mice. T regulatory cells (Tregs) could mediate the escape of T. crassiceps from the host immunity. This study is aimed to investigate the role of Tregs in T. crassiceps establishment in susceptible and non-susceptible mouse strains. Treg and effector cells were quantified in lymphoid organs before infection and 5, 30, 90, and 130 days post-infection. The proliferative response post-infection was characterized in vitro. The expression of regulatory and inflammatory molecules was assessed on days 5 and 30 post-infection. Depletion assays were performed to assess Treg functionality. Significantly higher Treg percentages were observed in BALB/cAnN mice, while increased percentages of activated CD127+ cells were found in C57BL/6 mice. The proliferative response was suppressed in susceptible mice, and Treg proliferation occurred only in susceptible mice. Treg-mediated suppression mechanisms may include IL-10 and TGFβ secretion, granzyme- and perforin-mediated cytolysis, metabolic disruption, and cell-to-cell contact. Tregs are functional in BALB/cAnN mice. Therefore Tregs could be allowing parasite establishment and survival in susceptible mice but could play a homeostatic role in non-susceptible strains.
Collapse
Affiliation(s)
- Laura Adalid-Peralta
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico.,Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, Mexico
| | | | - Cynthia Camacho-Vázquez
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Marisol Nájera-Ocampo
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Adrián Guevara-Salinas
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Nataly Ruiz-Monroy
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Marlene Melo-Salas
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Valeria Morales-Ruiz
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Dina López-Recinos
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Edgar Ortiz-Hernández
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | | | - Joel A Vazquez-Perez
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan, Mexico
| | - Asiel Arce-Sillas
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | - Sandra Gomez-Fuentes
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de la UNAM en el Instituto Nacional de Neurología y Neurocirugía México, Ciudad de México, Mexico
| | | | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Edda Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Edgar E Sevilla-Reyes
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan, Mexico
| |
Collapse
|
6
|
Yamamoto K, Makino T, Sato E, Noma T, Urakawa S, Takeoka T, Yamashita K, Saito T, Tanaka K, Takahashi T, Kurokawa Y, Yamasaki M, Nakajima K, Mori M, Doki Y, Wada H. Tumor-infiltrating M2 macrophage in pretreatment biopsy sample predicts response to chemotherapy and survival in esophageal cancer. Cancer Sci 2020; 111:1103-1112. [PMID: 31981293 PMCID: PMC7156837 DOI: 10.1111/cas.14328] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
The association between the tumor microenvironment (TME) and treatment response or survival has been a recent focus in several types of cancer. However, most study materials are resected specimens that were completely modified by prior chemotherapy; therefore, the unmodified host immune condition has not yet been clarified. The aim of the present study was to evaluate the relationship between TME assessed in pre-therapeutic biopsy samples and chemoresistance in esophageal cancer (EC). A total of 86 endoscopic biopsy samples from EC patients who received neoadjuvant chemotherapy (NAC) prior to surgery were evaluated for the number of intratumoral CD4+ lymphocytes (with/without Foxp3 expression), CD8+ lymphocytes (with/without PD-1 expression), monocytes (CD14+ ) and macrophages (CD86+ , CD163+ and CD206+ ) by multiplex immunohistochemistry (IHC). The number of tumor-infiltrating CD206+ macrophages I significantly correlated with cT, cM, cStage and neutrophil/lymphocyte ratio (NLR), whereas the number of lymphocytes (including expression of Foxp3 and PD-1) was not associated with clinico-pathological features. The high infiltration of CD163+ or CD206+ macrophages was significantly associated with poor pathological response to NAC (P = 0.0057 and 0.0196, respectively). Expression of arginase-1 in CD163+ macrophages tended to be higher in non-responders (29.4% vs 18.2%, P = 0.17). In addition, patients with high infiltration of M2 macrophages exhibited unfavorable overall survival compared to those without high infiltration of M2 macrophages (5-year overall survival 57.2% vs 71.0%, P = 0.0498). Thus, a comprehensive analysis of TME using multiplex IHC revealed that M2 macrophage infiltration would be useful in predicting the response to NAC and long-term survival in EC patients.
Collapse
Affiliation(s)
- Kei Yamamoto
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
- Department of Clinical Research in Tumor ImmunologyOsaka University Graduate School of MedicineOsakaJapan
| | - Tomoki Makino
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Eiichi Sato
- Department of PathologyInstitute of Medical Science (Medical Research Center)Tokyo Medical UniversityTokyoJapan
| | - Toshiki Noma
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Shinya Urakawa
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
- Department of Clinical Research in Tumor ImmunologyOsaka University Graduate School of MedicineOsakaJapan
| | - Tomohira Takeoka
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
- Department of Clinical Research in Tumor ImmunologyOsaka University Graduate School of MedicineOsakaJapan
| | - Kotaro Yamashita
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Takuro Saito
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Koji Tanaka
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Yukinori Kurokawa
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Makoto Yamasaki
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Kiyokazu Nakajima
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Masaki Mori
- Department of Surgery and ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Hisashi Wada
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversityOsakaJapan
- Department of Clinical Research in Tumor ImmunologyOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
7
|
Improved Anti-Treg Vaccination Targeting Foxp3 Efficiently Decreases Regulatory T Cells in Mice. J Immunother 2017; 39:269-75. [PMID: 27404943 DOI: 10.1097/cji.0000000000000133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The critical role of regulatory T (Treg) cells in dampening immune responses against tumor cells is apparent. Therefore, several methods have been introduced for eliminating Treg. Among them, inducing immune responses against Treg cells expressing Foxp3 transcription factor is a hopeful approach to decrease the frequency of Tregs. In current study, we used the chimeric FoxP3-Fc(IgG) fusion construct/protein to effectively stimulate the immune responses against Treg cells. MATERIALS AND METHODS Previously constructed FoxP3-Fc(IgG) DNA vaccine and its protein counterpart were injected into C57BL/6 mice in a prime/boost regimen. After 2 weeks, the mice were killed to measure the frequency of Tregs in their spleens, as well as analyze their specific cytokine production, T-cell proliferation, and CD8 T-cell cytotoxicity against FoxP3 protein. RESULTS FACS analysis of FoxP3 CD4 cells in splenocytes revealed the efficiency of FoxP3 DNA-prime protein-boost strategy to decrease the Treg cells and further showed considerable superiority of Fc(IgG) fusion strategy. This significant reduction in Treg frequency was also concomitant with higher FoxP3-specific CTL and Th1 responses in FoxP3-Fc vaccinated animals. CONCLUSIONS Prime/boost vaccination against FoxP3 in addition to enhanced antigen presentation by means of Fc fusion strategy could be successfully considered for Treg depletion studies. Validity of this approach should be experimentally tested in preclinical tumor models.
Collapse
|
8
|
Molecular adjuvants that modulate regulatory T cell function in vaccination: A critical appraisal. Pharmacol Res 2017; 129:237-250. [PMID: 29175113 DOI: 10.1016/j.phrs.2017.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/13/2022]
Abstract
Adjuvants are substances used to enhance the efficacy of vaccines. They influence the magnitude and alter the quality of the adaptive immune response to vaccine antigens by amplifying or modulating different signals involved in the innate immune response. The majority of known adjuvants have been empirically identified. The limited immunogenicity of new vaccine antigens and the need for safer vaccines have increased the importance of identifying single, well-defined adjuvants with known cellular and molecular mechanisms for rational vaccine design. Depletion or functional inhibition of CD4+CD25+FoxP3+ regulatory T cells (Tregs) by molecular adjuvants has become an emergent approach in this field. Different successful results have been obtained for specific vaccines, but there are still unresolved issues such as the risk of autoimmune disease induction, the involvement of cells other than Tregs and optimization for different conditions. This work provides a comprehensive analysis of current approaches to inhibit Tregs with molecular adjuvants for vaccine improvement, highlights the progress being made, and describes ongoing challenges.
Collapse
|
9
|
Development of a Curative Therapeutic Vaccine (TheraVac) for the Treatment of Large Established Tumors. Sci Rep 2017; 7:14186. [PMID: 29079801 PMCID: PMC5660153 DOI: 10.1038/s41598-017-14655-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/16/2017] [Indexed: 01/12/2023] Open
Abstract
Harnessing immune system to treat cancer requires simultaneous generation of tumor-specific CTLs and curtailment of tumor immunosuppressive environment. Here, we developed an immunotherapeutic regimen capable of eliminating large established mouse tumors using HMGN1, a DC-activating TLR4 agonist capable of inducing anti-tumor immunity. Intratumoral delivery of HMGN1 with low dose of Cytoxan cured mice bearing small (∅ ≈ 0.5 cm), but not large (∅ ≈ 1.0 cm) CT26 tumors. Screening for activators capable of synergizing with HMGN1 in activating DC identified R848. Intratumoral delivery of HMGN1 and R848 plus Cytoxan eradicated large established CT26 tumors. The resultant tumor-free mice were resistant to subsequent challenge with CT26, indicating the generation of CT26-specific protective immunity. This immunotherapeutic regimen caused homing of tumor-infiltrating DC to draining lymph nodes and increased infiltration of T cells into tumor tissues. Cytoxan in this regimen could be replaced by anti-CTLA4) or anti-PD-L1. Importantly, this immunotherapeutic regimen was also curative for large established mouse Renca and EG7 tumors. Thus, we have developed a curative therapeutic vaccination regimen dubbed ‘TheraVac’ consisting of HMGN1 and R848 plus a checkpoint inhibitor, that can, without using exogenous tumor-associated antigen(s), eliminate various large tumors and induce tumor-specific immunity.
Collapse
|
10
|
Han Z, Yang D, Trivett A, Oppenheim JJ. Therapeutic vaccine to cure large mouse hepatocellular carcinomas. Oncotarget 2017; 8:52061-52071. [PMID: 28881713 PMCID: PMC5581012 DOI: 10.18632/oncotarget.19367] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/10/2017] [Indexed: 01/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide with limited therapeutic options. Here we report the development of a therapeutic vaccination regimen (shortened as ‘TheraVac’) consisting of intratumoral delivery of high-mobility group nucleosome-binding protein 1 (HMGN1), R848/resiquimod, and one of the checkpoint inhibitors (e.g. anti-CTLA4, anti-PD-L1, or low dose of Cytoxan). C57BL/6 mice harboring large (approximately 1 cm in diameter) established subcutaneous Hepa1-6 hepatomas were cured by intratumoral injections of TheraVac and became tumor-free long-term survivors. Importantly, the resultant tumor-free mice were resistant to re-challenge with Hepa1-6 hepatoma, not B16 melanoma, demonstrating the acquisition of hepatoma-specific immunity in the absence of any administered tumor antigen. Mechanistic studies showed that upon treatment with TheraVac, Hepa1-6-bearing mice generated increased Hepa1-6-specific CTLs in the draining lymph nodes and showed greatly upregulated expression of CXCL9, CXCL10, and IFN-γ and elevated infiltration of T lymphocytes in tumor tissues. Treatment of large Hepa1-6 hepatomas on one mouse flank also eliminated smaller (approximately 0.5 cm in diameter) hepatomas implanted on the other flank. Thus, TheraVac has potential as a curative immunotherapeutic regimen for the treatment of human HCC.
Collapse
Affiliation(s)
- Zhen Han
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - De Yang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Anna Trivett
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Joost J Oppenheim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
11
|
IL-10: A Multifunctional Cytokine in Viral Infections. J Immunol Res 2017; 2017:6104054. [PMID: 28316998 PMCID: PMC5337865 DOI: 10.1155/2017/6104054] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/13/2017] [Accepted: 02/01/2017] [Indexed: 12/15/2022] Open
Abstract
The anti-inflammatory master regulator IL-10 is critical to protect the host from tissue damage during acute phases of immune responses. This regulatory mechanism, central to T cell homeostasis, can be hijacked by viruses to evade immunity. IL-10 can be produced by virtually all immune cells, and it can also modulate the function of these cells. Understanding the effects of this multifunctional cytokine is therefore a complex task. In the present review we discuss the factors driving IL-10 production and the cellular sources of the cytokine during antiviral immune responses. We particularly focus on the IL-10 regulatory mechanisms that impact antiviral immune responses and how viruses can use this central regulatory pathway to evade immunity and establish chronic/latent infections.
Collapse
|
12
|
Park JH, Jang M, Tarhan YE, Katagiri T, Sasa M, Miyoshi Y, Kalari KR, Suman VJ, Weinshilboum R, Wang L, Boughey JC, Goetz MP, Nakamura Y. Clonal expansion of antitumor T cells in breast cancer correlates with response to neoadjuvant chemotherapy. Int J Oncol 2016; 49:471-8. [PMID: 27278091 PMCID: PMC4922832 DOI: 10.3892/ijo.2016.3540] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/20/2016] [Indexed: 11/06/2022] Open
Abstract
The immune microenvironment of tumor plays a critical role in therapeutic responses to chemotherapy. Cancer tissues are composed of a complex network between antitumor and pro-tumor immune cells and molecules; therefore a comprehensive analysis of the tumor immune condition is imperative for better understanding of the roles of the immune microenvironment in anticancer treatment response. In this study, we performed T cell receptor (TCR) repertoire analysis of tumor infiltrating T cells (TILs) in cancer tissues of pre- and post-neoadjuvant chemotherapy (NAC) from 19 breast cancer patients; five cases showed CR (complete response), ten showed PR (partial response), and four showed SD/PD (stable disease/progressive disease) to the treatment. From the TCR sequencing results, we calculated the diversity index of the TCRβ chain and found that clonal expansion of TILs could be detected in patients who showed CR or PR to NAC. Noteworthy, the diversity of TCR was further reduced in the post-NAC tumors of CR patients. Our quantitative RT-PCR also showed that expression ratio of CD8/Foxp3 was significantly elevated in the post-NAC tumors of CR cases (p=0.0032), indicating that antitumor T cells were activated and enriched in these tumors. Collectively, our findings suggest that the clonal expansion of antitumor T cells may be a critical factor associated with response to chemotherapy and that their TCR sequences might be applicable for the development of TCR-engineered T cells treatment for individual breast cancer patients when their tumors relapse.
Collapse
Affiliation(s)
- Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Miran Jang
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Yunus Emre Tarhan
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Toyomasa Katagiri
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima 770-8503, Japan
| | - Mitsunori Sasa
- Department of Surgery, Tokushima Breast Care Clinic, Tokushima 770-0052, Japan
| | - Yasuo Miyoshi
- Division of Breast and Endocrine Surgery, Department of Surgery, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Krishna R. Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Vera J. Suman
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Judy C. Boughey
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew P. Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
13
|
Wang S, Zhang Y, Wang Y, Ye P, Li J, Li H, Ding Q, Xia J. Amphiregulin Confers Regulatory T Cell Suppressive Function and Tumor Invasion via the EGFR/GSK-3β/Foxp3 Axis. J Biol Chem 2016; 291:21085-21095. [PMID: 27432879 DOI: 10.1074/jbc.m116.717892] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Indexed: 11/06/2022] Open
Abstract
Previous studies mainly focused on the role of the epidermal growth factor receptor (EGFR) in tumor cells, whereas the effects of the EGFR on immune responses has not been determined. Our study shows that the EGFR signaling pathway play a role in the regulation of regulatory T cells (Treg cells) in cancer patients. The EGF-like growth factor Amphiregulin (AREG) protein was frequently up-regulated in a tissue microarray, which was associated with worse overall survival. Additionally, in sera, tissue specimens, and effusions of lung or gastric cancer patients, up-regulated AREG protein enhanced the suppressive function of Treg cells. AREG maintained the Treg cell suppressive function via the EGFR/GSK-3β/Foxp3 axis in vitro and in vivo Furthermore, inhibition of EGFR by the tyrosine kinase inhibitor gefitinib restored the activity of GSK-3β and attenuated Treg cell function. β-TrCP was involved in GSK-3β-mediated Foxp3 degradation, and mass spectrometry identified Lys356 as the ubiquitination site of Foxp3 by β-TrCP. These findings demonstrate the posttranslational regulation of Foxp3 expression by AREG in cancer patients through AREG/EGFR/GSK-3β signaling, which could lead to Foxp3 protein degradation in Treg cells and a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Sihua Wang
- From the Departments of Thoracic Surgery
| | | | - Yan Wang
- the Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, and
| | - Ping Ye
- the Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan 430022, China
| | - Jun Li
- the Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan 430022, China
| | - Huabin Li
- the Department of Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Qingqing Ding
- the Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, and
| | - Jiahong Xia
- Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,
| |
Collapse
|
14
|
Beyond adjuvants: Antagonizing inflammation to enhance vaccine immunity. Vaccine 2016; 33 Suppl 2:B55-9. [PMID: 26022570 DOI: 10.1016/j.vaccine.2015.03.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 11/20/2022]
Abstract
Since the development of the first vaccine over 200 years ago, vaccines have saved millions of lives and have become the most cost-effective modern medical intervention. However, over 70 years ago, Freund recognized that the effectiveness of the vaccine-induced immune responses could be vastly improved via the co-delivery of inflammation-induced agents, giving birth to the adjuvant field. Since the first description of adjuvants, revolutionary discoveries, including the discovery of dendritic cells and pattern recognition receptors, that drive remarkably different biological profiles, have opened the landscape of opportunities for the development of novel adjuvants able to trigger a remarkably diverse inflammatory profiles, thereby qualitatively and quantitatively skewing adaptive immunity in a tailored manner against target pathogens. However, mounting data point to a critical role for pre-existing inflammation as a predictor of vaccine responsiveness. Thus, in this review we will discuss novel opportunities by which pre-existing inflammation may be modulated, skewed, or tuned via next-generation vaccine approaches to enhanced vaccine-induced immunity in the elderly, immunocompromised, or subjects with chronic diseases.
Collapse
|
15
|
Pilon-Thomas S, Kodumudi KN, El-Kenawi AE, Russell S, Weber AM, Luddy K, Damaghi M, Wojtkowiak JW, Mulé JJ, Ibrahim-Hashim A, Gillies RJ. Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res 2015; 76:1381-90. [PMID: 26719539 DOI: 10.1158/0008-5472.can-15-1743] [Citation(s) in RCA: 448] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/09/2015] [Indexed: 12/21/2022]
Abstract
Cancer immunotherapies, such as immune checkpoint blockade or adoptive T-cell transfer, can lead to durable responses in the clinic, but response rates remain low due to undefined suppression mechanisms. Solid tumors are characterized by a highly acidic microenvironment that might blunt the effectiveness of antitumor immunity. In this study, we directly investigated the effects of tumor acidity on the efficacy of immunotherapy. An acidic pH environment blocked T-cell activation and limited glycolysis in vitro. IFNγ release blocked by acidic pH did not occur at the level of steady-state mRNA, implying that the effect of acidity was posttranslational. Acidification did not affect cytoplasmic pH, suggesting that signals transduced by external acidity were likely mediated by specific acid-sensing receptors, four of which are expressed by T cells. Notably, neutralizing tumor acidity with bicarbonate monotherapy impaired the growth of some cancer types in mice where it was associated with increased T-cell infiltration. Furthermore, combining bicarbonate therapy with anti-CTLA-4, anti-PD1, or adoptive T-cell transfer improved antitumor responses in multiple models, including cures in some subjects. Overall, our findings show how raising intratumoral pH through oral buffers therapy can improve responses to immunotherapy, with the potential for immediate clinical translation.
Collapse
Affiliation(s)
- Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Krithika N Kodumudi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Asmaa E El-Kenawi
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Shonagh Russell
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amy M Weber
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kimberly Luddy
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mehdi Damaghi
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jonathan W Wojtkowiak
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - James J Mulé
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Arig Ibrahim-Hashim
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
16
|
Regulatory T cells in the immunotherapy of melanoma. Tumour Biol 2015; 37:77-85. [PMID: 26515336 DOI: 10.1007/s13277-015-4315-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022] Open
Abstract
Patients with melanoma are supposed to develop spontaneous immune responses against specific tumor antigens. However, several mechanisms contribute to the failure of tumor antigen-specific T cell responses, inducing immune escape. Importantly, immunosuppression mediated by regulatory T cells (Tregs) in tumor lesions is a dominant mechanism of tumor immune evasion. Based on this information, several therapies targeting Tregs such as cyclophosphamide, IL-2-based therapies, and antibodies against the surface molecular of Tregs have been developed. However, only some of these strategies showed clinical efficacy in patients with melanoma in spite of their success in shifting immune systems to antitumor responses in animal models. In the future, strategies specifically depleting local Tregs, inhibiting Treg migration to the tumor lesion, and Treg depletion in combination with other chemotherapies or immune modulation will hopefully bring benefits to melanoma patients.
Collapse
|
17
|
Karpanen T, Olweus J. T-cell receptor gene therapy--ready to go viral? Mol Oncol 2015; 9:2019-42. [PMID: 26548533 DOI: 10.1016/j.molonc.2015.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/16/2022] Open
Abstract
T lymphocytes can be redirected to recognize a tumor target and harnessed to combat cancer by genetic introduction of T-cell receptors of a defined specificity. This approach has recently mediated encouraging clinical responses in patients with cancers previously regarded as incurable. However, despite the great promise, T-cell receptor gene therapy still faces a multitude of obstacles. Identification of epitopes that enable effective targeting of all the cells in a heterogeneous tumor while sparing normal tissues remains perhaps the most demanding challenge. Experience from clinical trials has revealed the dangers associated with T-cell receptor gene therapy and highlighted the need for reliable preclinical methods to identify potentially hazardous recognition of both intended and unintended epitopes in healthy tissues. Procedures for manufacturing large and highly potent T-cell populations can be optimized to enhance their antitumor efficacy. Here, we review the current knowledge gained from preclinical models and clinical trials using adoptive transfer of T-cell receptor-engineered T lymphocytes, discuss the major challenges involved and highlight potential strategies to increase the safety and efficacy to make T-cell receptor gene therapy a standard-of-care for large patient groups.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| |
Collapse
|
18
|
Cho HI, Jung SH, Sohn HJ, Celis E, Kim TG. An optimized peptide vaccine strategy capable of inducing multivalent CD8 + T cell responses with potent antitumor effects. Oncoimmunology 2015; 4:e1043504. [PMID: 26451316 PMCID: PMC4589052 DOI: 10.1080/2162402x.2015.1043504] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
Therapeutic cancer vaccines are an attractive alternative to conventional therapies for treating malignant tumors, and successful tumor eradication depends primarily on obtaining high numbers of long-lasting tumor-reactive CD8+ T cells. Dendritic cell (DC)-based vaccines constitute a promising approach for treating cancer, but in most instances low immune responses and suboptimal therapeutic effects are achieved indicating that further optimization is required. We describe here a novel vaccination strategy with peptide-loaded DCs followed by a mixture of synthetic peptides, polyinosine-polycytidylic acid (poly-IC) and anti-CD40 antibodies (TriVax) for improving the immunogenicity and therapeutic efficacy of DC-based vaccines in a melanoma mouse model. TriVax immunization 7–12 d after priming with antigen-loaded DCs generated large numbers of long-lasting multiple antigen-specific CD8+ T cells capable of recognizing tumor cells. These responses were far superior to those generated by homologous immunizations with either TriVax or DCs. CD8+ T cells but not CD4+ T cells or NK cells mediated the therapeutic efficacy of this heterologous prime-boost strategy. Moreover, combinations of this vaccination regimen with programmed cell death-1 (PD-1) blockade or IL2 anti-IL2 antibody complexes led to complete disease eradication and survival enhancement in melanoma-bearing mice. The overall results suggest that similar strategies would be applicable for the design of effective therapeutic vaccination for treating viral diseases and various cancers, which may circumvent current limitations of cell-based cancer vaccines.
Collapse
Affiliation(s)
- Hyun-Il Cho
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Soo-Hyun Jung
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Hyun-Jung Sohn
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Esteban Celis
- Cancer Immunology; Inflammation and Tolerance Program; Georgia Regents University Cancer Center ; Augusta, GA USA
| | - Tai-Gyu Kim
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; College of Medicine; The Catholic University of Korea ; Seoul, South Korea
| |
Collapse
|
19
|
Gao X, Wang X, Yang Q, Zhao X, Wen W, Li G, Lu J, Qin W, Qi Y, Xie F, Jiang J, Wu C, Zhang X, Chen X, Turnquist H, Zhu Y, Lu B. Tumoral expression of IL-33 inhibits tumor growth and modifies the tumor microenvironment through CD8+ T and NK cells. THE JOURNAL OF IMMUNOLOGY 2014; 194:438-45. [PMID: 25429071 DOI: 10.4049/jimmunol.1401344] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy has shown great promise as a new standard cancer therapeutic modality. However, the response rates are limited for current approach that depends on enhancing spontaneous antitumor immune responses. Therefore, increasing tumor immunogenicity by expressing appropriate cytokines should further improve the current immunotherapy. IL-33 is a member of the IL-1 family of cytokines and is released by necrotic epithelial cells or activated innate immune cells and is thus considered a "danger" signal. The role of IL-33 in promoting type 2 immune responses and tissue inflammation has been well established. However, whether IL-33 drives antitumor immune responses is controversial. Our previous work established that IL-33 promoted the function of CD8(+) T cells. In this study, we showed that the expression of IL-33 in two types of cancer cells potently inhibited tumor growth and metastasis. Mechanistically, IL-33 increased numbers and IFN-γ production by CD8(+) T and NK cells in tumor tissues, thereby inducing a tumor microenvironment favoring tumor eradication. Importantly, IL-33 greatly increased tumor Ag-specific CD8(+) T cells. Furthermore, both NK and CD8(+) T cells were required for the antitumor effect of IL-33. Moreover, depletion of regulatory T cells worked synergistically with IL-33 expression for tumor elimination. Our studies established "alarmin" IL-33 as a promising new cytokine for tumor immunotherapy through promoting cancer-eradicating type 1 immune responses.
Collapse
Affiliation(s)
- Xin Gao
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Xuefeng Wang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China; Department of Biochemistry and Molecular Biology, Soochow University, Suzhou 215007, People's Republic of China
| | - Qianting Yang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Key Laboratory of Infection and Immunity, Third People's Hospital, Guangdong Medical College, Shenzhen, Guangdong 518112, China
| | - Xin Zhao
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; The First Affiliated Hospital, Soochow University, Suzhou 215006, People's Republic of China
| | - Wen Wen
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Gang Li
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Junfeng Lu
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Haidian District, Beijing 100190, People's Republic of China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yuan Qi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Fang Xie
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Jingting Jiang
- The Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Changping Wu
- The Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Xueguang Zhang
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Xinchun Chen
- Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Key Laboratory of Infection and Immunity, Third People's Hospital, Guangdong Medical College, Shenzhen, Guangdong 518112, China
| | - Heth Turnquist
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and
| | - Yibei Zhu
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| |
Collapse
|
20
|
Jayaraman P, Alfarano MG, Svider PF, Parikh F, Lu G, Kidwai S, Xiong H, Sikora AG. iNOS expression in CD4+ T cells limits Treg induction by repressing TGFβ1: combined iNOS inhibition and Treg depletion unmask endogenous antitumor immunity. Clin Cancer Res 2014; 20:6439-51. [PMID: 25278453 DOI: 10.1158/1078-0432.ccr-13-3409] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Expression of inducible nitric oxide synthase (iNOS) in different cellular compartments may have divergent effects on immune function. We used a syngeneic tumor model to functionally characterize the role of iNOS in regulation of CD4(+)FOXP3(+) regulatory T cells (Treg), and optimize the beneficial effects of iNOS inhibition on antitumor immunity. EXPERIMENTAL DESIGN Wild-type (WT) or iNOS knockout mice bearing established MT-RET-1 melanoma were treated with the small-molecule iNOS inhibitor L-NIL and/or cyclophosphamide alone or in combination. The effect of iNOS inhibition or knockout on induction of Treg from mouse and human CD4(+) T cells in ex vivo culture was determined in parallel in the presence or absence of TGFβ1-depleting antibodies, and TGFβ1 levels were assessed by ELISA. RESULTS Whereas intratumoral myeloid-derived suppressor cells (MDSC) were suppressed by iNOS inhibition or knockout, systemic and intratumoral FOXP3(+) Treg levels increased in tumor-bearing mice. iNOS inhibition or knockout similarly enhanced induction of Treg from activated cultured mouse splenocytes or purified human or mouse CD4(+) T cells in a TGFβ1-dependent manner. Although either iNOS inhibition or Treg depletion with low-dose cyclophosphamide alone had little effect on growth of established MT-RET1 melanoma, combination treatment potently inhibited MDSC and Treg, boosted tumor-infiltrating CD8(+) T-cell levels, and arrested tumor growth in an immune-dependent fashion. CONCLUSIONS iNOS expression in CD4(+) T cells suppresses Treg induction by inhibiting TGFβ1 production. Our data suggest that iNOS expression has divergent effects on induction of myeloid and lymphoid-derived regulatory populations, and strongly support development of combinatorial treatment approaches that target these populations simultaneously.
Collapse
Affiliation(s)
- Padmini Jayaraman
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Dermatology, The Icahn School of Medicine at Mount Sinai, New York, New York. Immunology Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Head and Neck Cancer Research Program, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthew G Alfarano
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter F Svider
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Falguni Parikh
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Dermatology, The Icahn School of Medicine at Mount Sinai, New York, New York. Immunology Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Head and Neck Cancer Research Program, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Geming Lu
- Immunology Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah Kidwai
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Huabao Xiong
- Immunology Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York. Department of Dermatology, The Icahn School of Medicine at Mount Sinai, New York, New York. Immunology Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York. Head and Neck Cancer Research Program, The Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
21
|
Lykken JM, DiLillo DJ, Weimer ET, Roser-Page S, Heise MT, Grayson JM, Weitzmann MN, Tedder TF. Acute and chronic B cell depletion disrupts CD4+ and CD8+ T cell homeostasis and expansion during acute viral infection in mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:746-56. [PMID: 24928986 DOI: 10.4049/jimmunol.1302848] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
B cells provide humoral protection against pathogens and promote cellular immunity through diverse nonclassical effector functions. To assess B cell function in promoting T cell homeostasis, mature B cells were either acutely or chronically depleted in mice using CD20 mAb. Acute B cell depletion in either 2- or 4-mo-old mice significantly reduced spleen and lymph node CD4(+) and CD8(+) T cell numbers, including naive, activated, and Foxp3(+)CD25(+)CD4(+) regulatory T cell subsets. The numbers of IFN-γ- and TNF-α-producing T cells were also significantly reduced. Chronic B cell depletion for 6 mo in aged naive mice resulted in a 40-70% reduction in activated CD4(+) and CD8(+) T cell numbers and 20-50% reductions in IFN-γ-producing T cells. Therefore, B cells were necessary for maintaining naive CD4(+) and CD8(+) T cell homeostasis for subsequent optimal T cell expansion in young and old mice. To determine the significance of this finding, a week of B cell depletion in 4-mo-old mice was followed by acute viral infection with lymphocytic choriomeningitis virus Armstrong. Despite their expansion, activated and cytokine-producing CD4(+) and CD8(+) T cell numbers were still significantly reduced 1 wk later. Moreover, viral peptide-specific CD4(+) and CD8(+) T cell numbers and effector cell development were significantly reduced in mice lacking B cells, whereas lymphocytic choriomeningitis virus titers were dramatically increased. Thus, T cell function is maintained in B cell-depleted mice, but B cells are required for optimal CD4(+) and CD8(+) T cell homeostasis, activation, and effector development in vivo, particularly during responses to acute viral infection.
Collapse
Affiliation(s)
- Jacquelyn M Lykken
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - David J DiLillo
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Eric T Weimer
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Susanne Roser-Page
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033
| | - Mark T Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jason M Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157; and
| | - M Neale Weitzmann
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033; Division of Endocrinology, Metabolism and Lipids, Emory University School of Medicine, Atlanta, GA 30322
| | - Thomas F Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC 27710;
| |
Collapse
|
22
|
Rolinski J, Hus I. Breaking immunotolerance of tumors: a new perspective for dendritic cell therapy. J Immunotoxicol 2014; 11:311-8. [PMID: 24495309 DOI: 10.3109/1547691x.2013.865094] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The use of dendritic cells (DC) in cancer immunotherapy is based on their potent abilities to present antigens, so they can act as 'natural adjuvants' to enhance immunogenicity of tumor antigens and stimulate specific cytotoxic T-cells. Large amounts of DC can be generated from bone marrow, neonatal cord blood, and peripheral blood CD34(+) hematopoietic stem cells, or from peripheral blood monocytes. The DC can then be pulsed with tumor antigens and re-infused. In vitro, antigen-pulsed DC can stimulate allogeneic T-cell proliferation and induction of autologous specific cytotoxic T-cells; in vivo, the cells inhibit the growth of tumors or protect hosts (i.e. mice) from development of inoculated tumors. The results of preliminary clinical trials have shown that DC vaccines are safe and elicit immune responses; however, the rates of clinical responses are low. It has become quite clear that one key reason for unsatisfactory clinical results is tumor-induced immunosuppression. Among the factors contributing to this type of immunosuppression are populations of regulatory cells including: T-regulatory (T(reg)) cells, myeloid-derived suppressor cells (MDSC), tumor-associated macrophages (TAM), and DC expressing 2,3-dioxygenase indoleamine (IDO-DC). This review presents an overview of the current understanding about populations of regulatory cells and the most current research efforts directed to overcome immunosuppressive activity due to the tumor microenvironment.
Collapse
|
23
|
Bhopale MK, Hilliard B, Constantinescu CS, Fujioka T, Ventura E, Phillips SM, Rostami A. DAB389IL-2 suppresses autoimmune inflammation in the CNS and inhibits T cell-mediated lysis of glial target cells. Exp Mol Pathol 2014; 96:108-17. [DOI: 10.1016/j.yexmp.2013.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 07/10/2013] [Indexed: 12/20/2022]
|
24
|
Shimizu K, Kotera Y, Aruga A, Takeshita N, Katagiri S, Ariizumi SI, Takahashi Y, Yoshitoshi K, Takasaki K, Yamamoto M. Postoperative dendritic cell vaccine plus activated T-cell transfer improves the survival of patients with invasive hepatocellular carcinoma. Hum Vaccin Immunother 2014; 10:970-6. [PMID: 24419174 DOI: 10.4161/hv.27678] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The recurrence rate after surgery in patients with hepatocellular carcinoma (HCC) is very high, while prognosis is quite poor. However, there is no standard treatment to prevent recurrence of HCC after a curative operation. In this study, we investigated the clinical utilization of an autologous tumor lysate-pulsed dendritic cell vaccine plus ex vivo activated T cell transfer (ATVAC) in an adjuvant setting for postoperative HCC as a non-randomized controlled trial. Ninety-four patients with invasive HCC received informed consent information regarding the study, and 42 opted to have the ATVAC after surgery. Their recurrence-free survival (RFS) and overall survival (OS) were measured after 5 years and compared with those of 52 patients who selected to have the curative operation alone. The median RFS and OS were 24.5 months and 97.7 months in the patients receiving adjuvant ATVAC and 12.6 months and 41.0 months in the group receiving surgery alone (P = 0.011 and 0.029). In the treated group, patients with positive delayed-type hypersensitivity (DTH) had a better prognosis (RFS P = 0.019, OS P = 0.025). No adverse events of grade 3 or more were observed. A postoperative dendritic cell vaccine plus activated T cell transfer would be a feasible and effective treatment for preventing recurrence in HCC patients and achieving long-term survival especially in DTH positive patients.
Collapse
Affiliation(s)
- Koichi Shimizu
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Yoshihito Kotera
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Atsushi Aruga
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan; Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Nobuhiro Takeshita
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Satoshi Katagiri
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Shun-ichi Ariizumi
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Yutaka Takahashi
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Kenji Yoshitoshi
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Ken Takasaki
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| | - Masakazu Yamamoto
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
25
|
Wen M, Xu W, Ren L, Gao F, Cui N, Wen J, Li X, Lin L, Ma Z, Chen B, Cai J. Effector cells derived from naive T cells used in tumor immunotherapy of mice bearing B16 melanoma. Chin Med J (Engl) 2014; 127:1328-1333. [PMID: 24709189 DOI: 10.3760/cma.j.issn.0366-6999.20123364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adoptive cell transfer (ACT) immunotherapy has been used clinically for years to treat malignancies. Improving the killing efficiency of effector cells, such as tumor-specific cytotoxic T lymphocytes (CTLs), is an important component for enhancing the clinical response of cancer immunotherapy. Hence, we explored a novel method for preparing cancer-specific CTLs using naive T lymphocytes. METHODS C57BL/6 mice bearing B16 melanoma tumors were pretreated with cyclophosphamide (CTX) by peritoneal injection. The immunosuppressive influence of CTX on tumor regression and the tumor microenvironment was assessed. Naive T cells and T cell pools were isolated via negative selection using immunomagnetic beads. The proliferative potential and cytokine production of different T cell subpopulations were evaluated in vitro. Tumor-specific CTLs derived from naive T cells (naive CD4+ T cells: naive CD8+ T cells = 2:1) and pooled T cells were generated in vitro, respectively. B16 melanoma-bearing C57BL/6 mice were pretreated with CTX, followed by ACT immunotherapy using dendritic cell-induced CTLs. The homing abilities of the effector cells and interleukin-2 (IL-2), interferon-γ, granzyme B, and perforin mRNA levels in tumor tissues were evaluated, and the change in tumor volume was measured. RESULTS Mice receiving CTX peritoneal pretreatment injections did not display tumor regression compared with control mice. However, a significant downregulation of splenic Tregs and tumor growth factor-β1 (TGF-β1) and interleukin-10 (IL-10) serum levels was observed (P < 0.05). Naive T cells showed a stronger proliferative capacity and elevated cytokine production than did pooled T cells (P < 0.05). In addition, effector cells generated from naive T cells displayed more potent antitumor activity in vivo than those derived from pooled T cells (P < 0.05). CONCLUSION Effector cells derived from the naive T cells possess a stronger proliferative potential, homing capacity, and enhanced cytokine production, which leads to a superior antitumor response.
Collapse
Affiliation(s)
- Ming Wen
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, China; Department of Surgical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Weili Xu
- Department of Pediatric Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Lili Ren
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Fei Gao
- Teaching & Research Section, Hebei Medical University, Shijiazhuang, Hebei 050017; Department of Surgery, Department of Oncology & Immunotherapy, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Naipeng Cui
- Department of Surgical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Junye Wen
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Xinjiang Li
- Department of Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Lin Lin
- Department of Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Zhenfeng Ma
- Department of Surgical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Baoping Chen
- Department of Surgical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Jianhui Cai
- Teaching & Research Section, Hebei Medical University, Shijiazhuang, Hebei 050017; Department of Surgery, Department of Oncology & Immunotherapy, Hebei General Hospital, Shijiazhuang, Hebei 050051, China.
| |
Collapse
|
26
|
Okuyama R, Aruga A, Hatori T, Takeda K, Yamamoto M. Immunological responses to a multi-peptide vaccine targeting cancer-testis antigens and VEGFRs in advanced pancreatic cancer patients. Oncoimmunology 2013; 2:e27010. [PMID: 24498547 PMCID: PMC3906430 DOI: 10.4161/onci.27010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 02/06/2023] Open
Abstract
The prognosis of patients with advanced pancreatic cancer is extremely poor and there are only a few standard treatments. Here, we report the results of a Phase I clinical trial to investigate the safety, immunostimulatory effects, and antineoplastic activity of a multi-target vaccine composed of four distinct peptides derived from cancer-testis (CT) antigens and vascular endothelial growth factor receptors (VEGFRs). Nine patients with unresectable, advanced pancreatic cancer who were refractory to standard chemotherapy were enrolled. Each patient was vaccinated with HLA-A*2402-restricted peptides derived from the CT antigens kinesin family member 20A (KIF20A) and cell division cycle-associated 1 (CDCA1) as well as from VEGFR1 and VEGFR2 subcutaneously once a week, and disease progression was evaluated up to 6 mo later. Adverse events were assessed using the Common Terminology Criteria for Adverse Events v. 3.0. Immunological responses were monitored by ELISPOT assays and flow cytometry based on peptide-specific dextramers. The clinical outcomes that were measured were tumor response, progression-free survival (PFS) and overall survival (OS). In general, the multi-peptide vaccine was well-tolerated, and no grade 3 or 4 adverse events were observed upon vaccination. Peptide-specific T-cell responses were detected in all 9 patients, and clinical benefits were observed in four of them. Median PFS and OS were 90 and 207 d, respectively. The elicitation of multiple and robust peptide-specific T-cell responses as well as the status of host lymphocytes may be useful prognostic factors among patients with advanced pancreatic cancer treated with peptide-based anticancer vaccines.
Collapse
Affiliation(s)
- Ryuji Okuyama
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Tokyo, Japan
| | - Atsushi Aruga
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Tokyo, Japan ; Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Tokyo, Japan
| | - Takashi Hatori
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Tokyo, Japan
| | - Kazuyoshi Takeda
- Department of Immunology; Juntendo School of Medicine; Tokyo, Japan
| | - Masakazu Yamamoto
- Department of Gastroenterological Surgery; Tokyo Women's Medical University; Tokyo, Japan
| |
Collapse
|
27
|
Depletion of regulatory T cells by targeting folate receptor 4 enhances the potency of a GM-CSF-secreting tumor cell immunotherapy. Clin Immunol 2013; 148:287-98. [DOI: 10.1016/j.clim.2013.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/05/2013] [Accepted: 05/13/2013] [Indexed: 12/24/2022]
|
28
|
Tan C, Reddy V, Dannull J, Ding E, Nair SK, Tyler DS, Pruitt SK, Lee WT. Impact of anti-CD25 monoclonal antibody on dendritic cell-tumor fusion vaccine efficacy in a murine melanoma model. J Transl Med 2013; 11:148. [PMID: 23768240 PMCID: PMC3691646 DOI: 10.1186/1479-5876-11-148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022] Open
Abstract
Background A promising cancer vaccine involves the fusion of tumor cells with dendritic cells (DCs). As such, a broad spectrum of both known and unidentified tumor antigens is presented to the immune system in the context of the potent immunostimulatory capacity of DCs. Murine studies have demonstrated the efficacy of fusion immunotherapy. However the clinical impact of DC/tumor fusion vaccines has been limited, suggesting that the immunosuppresive milieu found in patients with malignancies may blunt the efficacy of cancer vaccination. Thus, novel strategies to enhance fusion vaccine efficacy are needed. Regulatory T cells (Tregs) are known to suppress anti-tumor immunity, and depletion or functional inactivation of these cells improves immunotherapy in both animal models and clinical trials. In this study, we sought to investigate whether functional inactivation of CD4+CD25+FoxP3+ Treg with anti-CD25 monoclonal antibody (mAb) PC61 prior to DC/tumor vaccination would significantly improve immunotherapy in the murine B16 melanoma model. Methods Treg blockade was achieved with systemic PC61 administration. This blockage was done in conjunction with DC/tumor fusion vaccine administration to treat established melanoma pulmonary metastases. Enumeration of these metastases was performed and compared between experimental groups using Wilcoxon Rank Sum Test. IFN-gamma ELISPOT assay was performed on splenocytes from treated mice. Results We demonstrate that treatment of mice with established disease using mAb PC61 and DC/tumor fusion significantly reduced counts of pulmonary metastases compared to treatment with PC61 alone (p=0.002) or treatment with control antibody plus fusion vaccine (p=0.0397). Furthermore, IFN-gamma ELISPOT analyses reveal that the increase in cancer immunity was mediated by anti-tumor specific CD4+ T-helper cells, without concomitant induction of CD8+ cytotoxic T cells. Lastly, our data provide proof of principle that combination treatment with mAb PC61 and systemic IL-12 can lower the dose of IL-12 necessary to obtain maximal therapeutic efficacy. Conclusions To our knowledge, this is the first report investigating the effects of anti-CD25 mAb administration on DC/tumor-fusion vaccine efficacy in a murine melanoma model, and our results may aide the design of future clinical trials with enhanced therapeutic impact.
Collapse
Affiliation(s)
- Chunrui Tan
- Division of Otolaryngology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ghansah T, Vohra N, Kinney K, Weber A, Kodumudi K, Springett G, Sarnaik AA, Pilon-Thomas S. Dendritic cell immunotherapy combined with gemcitabine chemotherapy enhances survival in a murine model of pancreatic carcinoma. Cancer Immunol Immunother 2013; 62:1083-91. [PMID: 23604104 DOI: 10.1007/s00262-013-1407-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 02/15/2013] [Indexed: 01/27/2023]
Abstract
Pancreatic cancer is an extremely aggressive malignancy with a dismal prognosis. Cancer patients and tumor-bearing mice have multiple immunoregulatory subsets including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC) that may limit the effectiveness of anti-tumor immunotherapies for pancreatic cancer. It is possible that modulating these subsets will enhance anti-tumor immunity. The goal of this study was to explore depletion of immunoregulatory cells to enhance dendritic cell (DC)-based cancer immunotherapy in a murine model of pancreatic cancer. Flow cytometry results showed an increase in both Tregs and MDSC in untreated pancreatic cancer-bearing mice compared with control. Elimination of Tregs alone or in combination with DC-based vaccination had no effect on pancreatic tumor growth or survival. Gemcitabine (Gem) is a chemotherapeutic drug routinely used for the treatment for pancreatic cancer patients. Treatment with Gem led to a significant decrease in MDSC percentages in the spleens of tumor-bearing mice, but did not enhance overall survival. However, combination therapy with DC vaccination followed by Gem treatment led to a significant delay in tumor growth and improved survival in pancreatic cancer-bearing mice. Increased MDSC were measured in the peripheral blood of patients with pancreatic cancer. Treatment with Gem also led to a decrease of this population in pancreatic cancer patients, suggesting that combination therapy with DC-based cancer vaccination and Gem may lead to improved treatments for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Tomar Ghansah
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Lee HJ, Seo JY, Ahn JH, Ahn SH, Gong G. Tumor-associated lymphocytes predict response to neoadjuvant chemotherapy in breast cancer patients. J Breast Cancer 2013; 16:32-9. [PMID: 23593079 PMCID: PMC3625767 DOI: 10.4048/jbc.2013.16.1.32] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 02/07/2013] [Indexed: 01/08/2023] Open
Abstract
Purpose Tumor-associated lymphocyte numbers in breast cancer have been suggested as a new independent predictor of response to neoadjuvant chemotherapy in breast cancer patients. We therefore evaluated the relationship between pathologic complete response (pCR) and tumor-associated lymphocytes in tumors of such patients. Methods Between 2000 and 2009, we retrospectively evaluated 175 patients with primary breast cancer treated with neoadjuvant chemotherapy, followed by definitive surgical resection. Peritumoral lymphocytic infiltration (LI) and CD3+, CD8+, and forkhead box P3 (FOXP3)+ lymphocytes were assessed in pretreatment biopsy specimens. Results Nineteen (11%) patients achieved pCR. An elevated LI, CD3+, CD8+, or FOXP3+ lymphocytic infiltration; lower clinical T stage; human epidermal growth factor receptor 2 overexpression; and herceptin-based treatment were all significantly associated with pCR. Through a multivariate analysis, LI (odds ratio [OR], 1.26; p=0.024), clinical T stage (OR, 3.06; p=0.041), and the use of a herceptin-based regimen (OR, 4.95; p=0.004) were all significant independent predictors of pCR. Significantly higher numbers of tumor-associated lymphocytes and CD3+, CD8+, and FOXP3+ T-cells were observed in the following: high-grade tumors, tumors of positive nodal status, and tumors negative for hormone receptors. Conclusion Tumor-associated lymphocytes are significantly associated with pCR, suggesting that tumor-associated lymphocytes may be an important pathological factor predicting a response to neoadjuvant chemotherapy in breast cancer patients.
Collapse
Affiliation(s)
- Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
31
|
Aruga A, Takeshita N, Kotera Y, Okuyama R, Matsushita N, Ohta T, Takeda K, Yamamoto M. Long-term Vaccination with Multiple Peptides Derived from Cancer-Testis Antigens Can Maintain a Specific T-cell Response and Achieve Disease Stability in Advanced Biliary Tract Cancer. Clin Cancer Res 2013; 19:2224-31. [PMID: 23479678 DOI: 10.1158/1078-0432.ccr-12-3592] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE The prognosis of patients with advanced biliary tract cancer (BTC) is extremely poor and there are only a few standard treatments. We conducted a phase I trial to investigate the safety, immune response, and antitumor effect of vaccination with four peptides derived from cancer-testis antigens, with a focus on their fluctuations during long-term vaccination until the disease had progressed. EXPERIMENTAL DESIGN Nine patients with advanced BTC who had unresectable tumors and were refractory to standard chemotherapy were enrolled. HLA-A*2402-restricted epitope peptides, lymphocyte antigen 6 complex locus K, TTK protein kinase, insulin-like growth factor-II mRNA-binding protein 3, and DEP domain containing 1 were vaccinated subcutaneously once a week at doses of 0.5, 1, or 2 mg and continued until disease progression. The adverse events were assessed by Common Terminology Criteria for Adverse Events and the immune response was monitored by an enzyme-linked immunospot assay or by flow cytometry. The clinical effects observed were tumor response, progression-free survival (PFS), and overall survival (OS). RESULTS Four-peptide vaccination was well tolerated. No grade 3 or 4 adverse events were observed. Peptide-specific T-cell immune responses were observed in seven of nine patients and clinical responses were observed in six of nine patients. The median PFS and OS were 156 and 380 days. The injection site reaction and CTL induction seemed to be prognostic factors of both PFS and OS. CONCLUSIONS Four-peptide vaccination was well tolerated and seemed to provide some clinical benefit to some patients. These immunologic and clinical responses were maintained over the long term through continuous vaccinations.
Collapse
Affiliation(s)
- Atsushi Aruga
- Department of Gastroenterological Surgery; Institute of Advanced Biomedical Engineering and Science, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Zaiss DMW, van Loosdregt J, Gorlani A, Bekker CPJ, Gröne A, Sibilia M, van Bergen en Henegouwen PMP, Roovers RC, Coffer PJ, Sijts AJAM. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor. Immunity 2013; 38:275-84. [PMID: 23333074 PMCID: PMC3582723 DOI: 10.1016/j.immuni.2012.09.023] [Citation(s) in RCA: 331] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/27/2012] [Indexed: 12/16/2022]
Abstract
Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients.
Collapse
Affiliation(s)
- Dietmar M W Zaiss
- Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, University of Utrecht, 3584 CL Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zaiss DMW, van Loosdregt J, Gorlani A, Bekker CPJ, Gröne A, Sibilia M, van Bergen en Henegouwen PMP, Roovers RC, Coffer PJ, Sijts AJAM. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor. Immunity 2013. [PMID: 23333074 DOI: 10.1016/j.immuni.2012.09.023.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients.
Collapse
Affiliation(s)
- Dietmar M W Zaiss
- Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, University of Utrecht, 3584 CL Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chopra M, Riedel SS, Biehl M, Krieger S, von Krosigk V, Bäuerlein CA, Brede C, Jordan Garrote AL, Kraus S, Schäfer V, Ritz M, Mattenheimer K, Degla A, Mottok A, Einsele H, Wajant H, Beilhack A. Tumor necrosis factor receptor 2-dependent homeostasis of regulatory T cells as a player in TNF-induced experimental metastasis. Carcinogenesis 2013; 34:1296-303. [PMID: 23385062 DOI: 10.1093/carcin/bgt038] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cytokine tumor necrosis factor (TNF) has pleiotropic functions both in normal physiology and disease. TNF signals by the virtue of two cell surface receptors, TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Exogenous TNF promotes experimental metastasis in some models, yet the underlying mechanisms are poorly understood. To study the contribution of host TNFR1 and TNFR2 on tumor cell progression and metastasis, we employed a syngeneic B16F10 melanoma mouse model of lung metastasis combined with in vivo bioluminescence imaging. Treatment of tumor-bearing mice with recombinant human TNF resulted in a significant increase in tumor burden and metastatic foci. This correlated with an increase in pulmonary regulatory CD4(+)/Foxp3(+) T cells. TNF caused an expansion of regulatory T (Treg) cells in vitro in a TNFR2-dependent manner. To assess the contribution of immune cell expression of endogenous TNF and its two receptors on B16F10 metastasis, we generated bone marrow chimeras by reconstituting wild-type mice with bone marrow from different knockout mice. Loss of either TNF or TNFR2 on immune cells resulted in decreased B16F10 metastasis and lower numbers of Treg cells within the lungs of these animals. Selective depletion of Treg cells attenuated metastasis even in conjunction with TNF treatment. We propose a novel mechanism in which TNF activates TNFR2 on Treg cells and thereby expands this immunosuppressive immune cell population. Loss of either TNF or TNFR2 prevents the accumulation of Treg cells and results in a less tolerogenic environment, enabling the immune system to control B16F10 tumor metastasis and growth.
Collapse
Affiliation(s)
- Martin Chopra
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oda N, Shimazu K, Naoi Y, Morimoto K, Shimomura A, Shimoda M, Kagara N, Maruyama N, Kim SJ, Noguchi S. Intratumoral regulatory T cells as an independent predictive factor for pathological complete response to neoadjuvant paclitaxel followed by 5-FU/epirubicin/cyclophosphamide in breast cancer patients. Breast Cancer Res Treat 2012; 136:107-16. [PMID: 22986814 DOI: 10.1007/s10549-012-2245-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/05/2012] [Indexed: 12/19/2022]
Abstract
Anti-tumor immunity is thought to play a significant role in chemotherapeutic response of breast tumors. In the present study, we investigated whether tumor infiltrating FOXP3+ regulatory T cells, CD8+ cytotoxic T cells, and IL17F+ helper T cells were associated with a pathological complete response (pCR) to neoadjuvant chemotherapy (NAC). Breast cancer patients (stages II and III, n = 180) who were treated with NAC consisting of sequential weekly paclitaxel followed by 5-FU/epirubicin/cyclophosphamide were included for this study. Core needle tumor specimens obtained before NAC were immunohistochemically examined for FOXP3, CD8, and IL17F. Intratumoral infiltration of FOXP3+, CD8+, and IL17F+ T cells was observed in 62.2, 80.0, and 62.2 % of tumors, respectively. FOXP3 and CD8 infiltrates, but not IL17F infiltrate, were significantly (P < 0.001 and P = 0.007, respectively) associated with a high-pCR rate (31.3 and 25.7 %, respectively), and breast tumors with both FOXP3 and CD8 infiltrates showed the highest pCR rate (33.0 %). Multivariate analysis indicated that only FOXP3 infiltrates (P = 0.014) and the conventional predictive factor Ki67 (P = 0.031) were statistically significant and independent predictors of pCR. Breast tumors with FOXP3 and CD8 infiltrates were more likely to achieve pCR. FOXP3 infiltrate, in combination with Ki67, could thus be used as a clinically useful predictor of response to NAC. The possible indirect mechanism through which chemotherapy exerts its anti-tumor activity, i.e., enhancing anti-tumor immunity by inhibiting FOXP3, was also suggested.
Collapse
Affiliation(s)
- Naofumi Oda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Facciabene A, Motz GT, Coukos G. T-regulatory cells: key players in tumor immune escape and angiogenesis. Cancer Res 2012; 72:2162-71. [PMID: 22549946 DOI: 10.1158/0008-5472.can-11-3687] [Citation(s) in RCA: 636] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
T-regulatory cells (Tregs) are found infiltrating tumors in a vast array of tumor types, and tumor-infiltrating Tregs are often associated with a poor clinical outcome. Tregs are potent immunosuppressive cells of the immune system that promote progression of cancer through their ability to limit antitumor immunity and promote angiogenesis. Here, we discuss the ways in which Tregs suppress the antitumor immune response and elaborate on our recent discovery that Tregs make significant direct contributions to tumor angiogenesis. Further, we highlight several current therapies aimed at eliminating Tregs in cancer patients. Given the multifaceted role of Tregs in cancer, a greater understanding of their functions will ultimately strengthen future therapies.
Collapse
Affiliation(s)
- Andrea Facciabene
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
37
|
Abstract
Many immunotherapeutic agents in phase II cancer studies have given optimistic results, which were not confirmed in larger randomized studies. Here we explore the evidence that, contrary to previous opinion, many chemotherapeutic agents and other classes of drugs may enhance the response to therapeutic vaccines by reducing inflammation and/or by inhibiting regulatory T lymphocytes or myeloid-derived suppressor cells. In addition, some of these agents, such as the immunomodulatory drugs, may produce marked costimulatory activities as in the case of lenalidomide, which also has marked anti-inflammatory properties. With the first approval for a vaccine-based therapy for prostate cancer, we propose that many more vaccines will be able to achieve approval, especially when combined with the optimal chemotherapy and/or immunomodulatory drug schedule.
Collapse
Affiliation(s)
- Wai M Liu
- Department of Oncology, Division of Clinical Sciences, St George's, University of London, London, UK
| | | |
Collapse
|
38
|
Wang X, Liu F, Zhou S, Xu Z, Hoellwarth J, Chen X, He L, Zhang R, Liu F, Wang J, Su C. Partial regulatory T cell depletion prior to schistosomiasis vaccination does not enhance the protection. PLoS One 2012; 7:e40359. [PMID: 22802961 PMCID: PMC3389001 DOI: 10.1371/journal.pone.0040359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 06/07/2012] [Indexed: 11/21/2022] Open
Abstract
CD4+CD25+ regulatory T cells (Tregs) do
not only influence self-antigen specific immune responses, but also dampen
the protective effect induced by a number of vaccines. The impact of CD4+CD25+
Tregs on vaccines against schistosomiasis, a neglected tropical disease that
is a major public health concern, however, has not been examined. In this
study, a DNA vaccine encoding a 26 kDa glutathione S-transferase of Schistosoma
japonicum (pVAX1-Sj26GST) was constructed and its potential effects
were evaluated by depleting CD25+ cells prior to pVAX1-Sj26GST
immunization. This work shows that removal of CD25+ cells
prior to immunization with the pVAX1-Sj26GST schistosomiasis DNA vaccine significantly
increases the proliferation of splenocytes and IgG levels. However, CD25+
cell-depleted mice immunized with pVAX1-Sj26GST show no improved protection
against S. japonicum. Furthermore, depletion of CD25+
cells causes an increase in both pro-inflammatory cytokines (e.g. IFN-γ,
GM-CSF and IL-4) and an anti-inflammatory cytokine (e.g. IL-10), with CD4+CD25-
T cells being one of the major sources of both IFN-γ and IL-10. These
findings indicate that partial CD25+ cell depletion fails
to enhance the effectiveness of the schistosome vaccine, possibly due to IL-10
production by CD4+CD25- T cells, or other cell
types, after CD25+ cell depletion during vaccination.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Pathogen Biology and Immunology, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Clinical utilization of postoperative dendritic cell vaccine plus activated T-cell transfer in patients with intrahepatic cholangiocarcinoma. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2012; 19:171-8. [PMID: 21874278 DOI: 10.1007/s00534-011-0437-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The prognosis of patients with intrahepatic cholangiocarcinoma (ICC) is extremely poor and the recurrence rate after curative operation is very high. There is no standard treatment to prevent recurrence of ICC. In this study, we investigated the clinical utilization of a dendritic cell vaccine plus activated T-cell transfer in an adjuvant setting for postoperative ICC. METHODS 36 patients with ICC were vaccinated at least 3 times with autologous tumor lysate pulsed dendritic cells plus ex-vivo activated T-cell transfer. The 5-year progression-free survival (PFS) and overall survival (OS) were measured and compared with those of 26 patients who received the curative operation alone as a concurrent control. The registration number was UMIN000005820. RESULTS The median PFS and OS were 18.3 and 31.9 months in the patients receiving adjuvant immunotherapy and 7.7 and 17.4 months in the group receiving surgery alone (p = 0.005 and 0.022, respectively). In the treated group, patients whose skin reactions were 3 cm or more at the vaccine site showed dramatically better prognosis (PFS p < 0.001, OS p = 0.001). CONCLUSIONS A postoperative dendritic cell vaccine plus activated T-cell transfer would be a feasible and effective treatment for preventing recurrence and achieving long-term survival in ICC patients.
Collapse
|
40
|
Jacobs JFM, Nierkens S, Figdor CG, de Vries IJM, Adema GJ. Regulatory T cells in melanoma: the final hurdle towards effective immunotherapy? Lancet Oncol 2012; 13:e32-42. [PMID: 22225723 DOI: 10.1016/s1470-2045(11)70155-3] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Immunotherapy studies in patients with melanoma have reported success in the expansion of tumour-specific effector T cells in vivo, but even in the presence of substantial numbers of functional T cells circulating in the blood, favourable clinical outcomes are scarce. This failure to induce robust clinical responses might be related to tumour-induced immune evasion, rendering the host tolerant to melanoma antigens. Immunosuppression in the tumour microenvironment mediated by regulatory T cells (Treg) is a dominant mechanism of tumour immune escape and is a major hurdle for tumour immunotherapy. Accumulation of Treg in melanoma is frequently recorded and the ratio of CD8-positive T cells versus Treg in the tumour microenvironment is predictive for survival of patients with melanoma. Hence, depletion of Treg seems to be a promising strategy for the enhancement of melanoma-specific immunity. Indeed, murine studies have shown that Treg depletion greatly increases the efficacy of immunotherapy. But despite the success of some strategies in depletion of Treg in patients, overall clinical efficacy has been disappointing. The lack of Treg specificity of the Treg depleting strategies applied so far imply that well-designed studies into dosage, timing, and administration regimens with more specific agents are urgently needed. Depletion of functional Treg from the tumour microenvironment as part of multifaceted immunotherapeutic treatments is a major challenge to induce clinically relevant immune responses against melanomas.
Collapse
Affiliation(s)
- Joannes F M Jacobs
- Department of Tumour Immunology, Nijmegen Centre for Molecular Life Sciences, Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
41
|
Mitchell L, Thamm D, Biller B. Clinical and Immunomodulatory Effects of Toceranib Combined with Low-Dose Cyclophosphamide in Dogs with Cancer. J Vet Intern Med 2012; 26:355-62. [DOI: 10.1111/j.1939-1676.2011.00883.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/02/2011] [Accepted: 12/19/2011] [Indexed: 12/16/2022] Open
Affiliation(s)
- L. Mitchell
- Animal Cancer Center; Department of Clinical Sciences; Colorado State University; Fort Collins; CO
| | - D.H. Thamm
- Animal Cancer Center; Department of Clinical Sciences; Colorado State University; Fort Collins; CO
| | - B.J. Biller
- Animal Cancer Center; Department of Clinical Sciences; Colorado State University; Fort Collins; CO
| |
Collapse
|
42
|
Knueppel A, Lange S, Altmann S, Sekora A, Knuebel G, Vogel H, Lindner I, Freund M, Junghanss C. Upfront Denileukin Diftitox as in vivo regulatory T-cell depletion in order to enhance vaccination effects in a canine allogeneic hematopoietic stem cell transplantation model. Vet Immunol Immunopathol 2012; 145:233-40. [DOI: 10.1016/j.vetimm.2011.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 11/11/2011] [Accepted: 11/14/2011] [Indexed: 01/01/2023]
|
43
|
Deciphering the Multifaceted Relationship between Oncolytic Viruses and Natural Killer Cells. Adv Virol 2011; 2012:702839. [PMID: 22312364 PMCID: PMC3263705 DOI: 10.1155/2012/702839] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/14/2011] [Indexed: 12/23/2022] Open
Abstract
Despite active research in virotherapy, this apparently safe modality has not achieved widespread success. The immune response to viral infection appears to be an essential factor that determines the efficacy of oncolytic viral therapy. The challenge is determining whether the viral-elicited immune response is a hindrance or a tool for viral treatment. NK cells are a key component of innate immunity that mediates antiviral immunity while also coordinating tumor clearance. Various reports have suggested that the NK response to oncolytic viral therapy is a critical factor in premature viral clearance while also mediating downstream antitumor immunity. As a result, particular attention should be given to the NK cell response to various oncolytic viral vectors and how their antiviral properties can be suppressed while maintaining tumor clearance. In this review we discuss the current literature on the NK response to oncolytic viral infection and how future studies clarify this intricate response.
Collapse
|
44
|
Rowswell-Turner RB, Harden JL, Nair RE, Gu T, Kilinc MO, Egilmez NK. Chronic chemoimmunotherapy achieves cure of spontaneous murine mammary tumors via persistent blockade of posttherapy counter-regulation. THE JOURNAL OF IMMUNOLOGY 2011; 187:4109-18. [PMID: 21908736 DOI: 10.4049/jimmunol.1101136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Intratumoral delivery of IL-12 and GM-CSF induces local and systemic antitumor CD8(+) T cell activation and tumor kill. However, the effector response is transient and is rapidly countered by CD4(+) Foxp3(+) T suppressor cell expansion. To determine whether depletion of the pre-existing T suppressor cell pool prior to treatment could diminish posttherapy regulatory cell resurgence, FVBneuN mice bearing advanced spontaneous mammary tumors were treated with cyclophosphamide (CY) 1 d before IL-12/GM-CSF therapy. Administration of CY mediated a significant delay in the post-IL-12/GM-CSF T suppressor cell rebound, resulting in a 7-fold increase in the CD8(+) CTL/T suppressor cell ratio, a 3-fold enhancement of CTL cytotoxicity, and an extension of the effector window from 3 to 7 d. In long-term therapy studies, chronic chemoimmunotherapy promoted a dramatic enhancement of tumor regression, resulting in complete cure in 44% of the mice receiving CY plus IL-12/GM-CSF. Tumor eradication in the chronic therapy setting was associated with the ability to repeatedly rescue and maintain cytotoxic CD8(+) T cell activity. These findings demonstrated that chronic administration of CY in conjunction with immune therapy enhances the initial induction of antitumor T effector cells and, more importantly, sustains their cytotoxic activity over the long-term via persistent blockade of homeostatic counter-regulation.
Collapse
Affiliation(s)
- Rachael B Rowswell-Turner
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214,USA
| | | | | | | | | | | |
Collapse
|
45
|
A CCR4 antagonist combined with vaccines induces antigen-specific CD8+ T cells and tumor immunity against self antigens. Blood 2011; 118:4853-62. [PMID: 21908423 DOI: 10.1182/blood-2011-01-329656] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) may impede cancer vaccine efficacy in hematologic malignancies and cancer. CCR4 antagonists, an emergent class of Treg inhibitor, have been shown to block recruitment of Tregs mediated by CCL22 and CCL17. Our aim was to demonstrate the ability of a CCR4 antagonist (a small chemical molecule identified in silico) when combined with vaccines to break peripheral tolerance controlled by Tregs, a prerequisite for the induction of CD8(+) T cells against self Ags. Immunization of transgenic or normal mice expressing tumor-associated self Ags (Her2/neu, OVA, gp100) with a CCR4 antagonist combined with various vaccines led to the induction of effector CD8(+) T cells and partial inhibition of tumor growth expressing self Ags in both prophylactic and therapeutic settings. The CCR4 antagonist was more efficient than cyclophosphamide to elicit anti-self CD8(+) T cells. We also showed that the population of Tregs expressing CCR4 corresponded to memory (CD44(high)) and activated (ICOS(+)) Tregs, an important population to be targeted to modulate Treg activity. CCR4 antagonist represents a competitive class of Treg inhibitor able to induce functional anti-self CD8(+) T cells and tumor growth inhibition when combined with vaccines. High expression of CCR4 on human Tregs also supports the clinical development of this strategy.
Collapse
|
46
|
Walczak M, Regts J, van Oosterhout AJM, Boon L, Wilschut J, Nijman HW, Daemen T. Role of regulatory T-cells in immunization strategies involving a recombinant alphavirus vector system. Antivir Ther 2011; 16:207-18. [PMID: 21447870 DOI: 10.3851/imp1751] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Regulatory T-cells (Treg) hamper immune responses elicited by cancer vaccines. Therefore, depletion of Treg is being used to improve the outcome of vaccinations. METHODS We studied whether an alphavirus vector-based immunotherapeutic vaccine changes the number and/or activity of Treg and if Treg depletion improves the efficacy of this vaccine against tumours. The vaccine is based on a Semliki Forest virus (SFV). The recombinant SFV replicon particles encode a fusion protein of E6 and E7 from human papillomavirus (HPV) type 16 (SFVeE6,7). RESULTS We demonstrated that SFVeE6,7 immunization did not change Treg levels and their suppressive activity. Depletion of Treg in mice, using the novel anti-folate receptor 4 antibody, did not enhance the immune response induced by SFVeE6,7 immunization. Both the priming and the proliferation phases of the HPV-specific response elicited with SFVeE6,7 were not affected by the immune-suppressive activity of Treg. Moreover, Treg depletion did not improve the therapeutic antitumour response of SFVeE6,7 in a murine tumour model. CONCLUSIONS The efficacy of the SFVeE6,7 vaccine was not hampered by Treg. Therefore, SFVeE6,7 seems a very promising candidate for the treatment of HPV-induced disease, as it may not require additional immune interventions to modulate Treg activity.
Collapse
Affiliation(s)
- Mateusz Walczak
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
47
|
Kato M, Nakamura Y, Suda T, Ozawa Y, Inui N, Seo N, Nagata T, Koide Y, Kalinski P, Nakamura H, Chida K. Enhanced anti-tumor immunity by superantigen-pulsed dendritic cells. Cancer Immunol Immunother 2011; 60:1029-38. [PMID: 21519830 PMCID: PMC11029592 DOI: 10.1007/s00262-011-1015-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 03/31/2011] [Indexed: 12/25/2022]
Abstract
Staphylococcal enterotoxins A (SEA) and B (SEB) are classical models of superantigens (SAg), which induce potent T-cell-stimulating activity by forming complexes with MHC class II molecules on antigen-presenting cells. This large-scale activation of T-cells is accompanied by increased production of cytokines such as interferon-γ (IFN-γ). Additionally, as we previously reported, IFN-γ-producing CD8(+) T cells act as "helper cells," supporting the ability of dendritic cells to produce interleukin-12 (IL-12)p70. Here, we show that DC pulsed with SAg promote the enhancement of anti-tumor immunity. Murine bone marrow-derived dendritic cells (DC) were pulsed with OVA(257-264) (SIINFEKL), which is an H-2Kb target epitope of EG7 [ovalbumin (OVA)-expressing EL4] cell lines, in the presence of SEA and SEB and were subcutaneously injected into naïve C57BL/6 mice. SAg plus OVA(257-264)-pulsed DC vaccine strongly enhanced peptide-specific CD8(+) T cells exhibiting OVA(257-264)-specific cytotoxic activity and IFN-γ production, leading to the induction of protective immunity against EG7 tumors. Furthermore, cyclophosphamide (CY) added to SAg plus tumor-antigens (OVA(257-264), tumor lysate, or TRP-2) pulsed DC immunization markedly enhanced tumor-specific T-cell expansion and had a significant therapeutic effect against various tumors (EG7, 2LL, and B16). Superantigens are potential candidates for enhancing tumor immunity in DC vaccines.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antineoplastic Agents, Alkylating/therapeutic use
- CD8-Positive T-Lymphocytes
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Cyclophosphamide/therapeutic use
- Cytokines/metabolism
- Dendritic Cells/immunology
- Flow Cytometry
- Histocompatibility Antigens Class II/metabolism
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lymphocyte Activation
- Lymphoma/drug therapy
- Lymphoma/immunology
- Lymphoma/metabolism
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Mice
- Mice, Inbred C57BL
- Ovalbumin/physiology
- Receptors, G-Protein-Coupled/physiology
- Superantigens/immunology
- Survival Rate
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Tumor Cells, Cultured
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Masato Kato
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yuichi Ozawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naohiro Seo
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Toshi Nagata
- Department of Health Science, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yukio Koide
- Department of Infectious Diseases, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Pawel Kalinski
- Department of Surgery, Immunology, Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA USA
| | - Hirotoshi Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Kingo Chida
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| |
Collapse
|
48
|
A potential role for CD25+ regulatory T-cells in the protection against casein allergy by dietary non-digestible carbohydrates. Br J Nutr 2011; 107:96-105. [DOI: 10.1017/s0007114511002637] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dietary non-digestible carbohydrates reduce the development of cows' milk allergy in mice. In the present study, the contribution of CD25+ regulatory T-cells (Treg) was investigated using in vivo Treg depletion and adoptive transfer studies. Mice were orally sensitised with casein and fed a diet containing 2 % short-chain galacto-, long-chain fructo- and acidic oligosaccharides (GFA) or a control diet. Donor splenocytes of mice sensitised with casein and fed the GFA or control diet were adoptively transferred to naive recipient mice, which were casein- or sham-sensitised and fed the control diet. In addition, in vivo or ex vivo CD25+ Treg depletion was performed using anti-CD25 (PC61). The acute allergic skin response upon intradermal casein challenge and casein-specific Ig were determined. Furthermore, T-helper (TH) 1 and TH2 cell numbers were analysed in the mesenteric lymph nodes. The oligosaccharide diet strongly reduced the development of the acute allergic skin response, which was abrogated by the in vivo anti-CD25 treatment. The diet enhanced the percentage of TH1 cells and tended to reduce the percentage of TH2 cells in casein-sensitised mice. Recipient mice were protected against the development of an acute allergic skin response when transferred with splenocytes from casein-sensitised GFA-fed donor mice before sensitisation. Ex vivo depletion of CD25+ Treg abrogated this transfer of tolerance. Splenocytes from sham-sensitised GFA-fed donor mice did not suppress the allergic response in recipient mice. In conclusion, CD25+ Treg contribute to the suppression of the allergic effector response in casein-sensitised mice induced by dietary intervention with non-digestible carbohydrates.
Collapse
|
49
|
Abstract
In this review, we introduce the changing public perception of vaccines and immunotherapy in cancer treatments. We discuss the roles that different immunosuppressive cells play in the tumor microenvironment. Tumor associated macrophages (TAMs) and M1 and M2 macrophage phenotypes are discussed in depth. Additionally, the role that myeloid derived suppressor cells (MDSC) and T regulatory cells (Tregs) play in the tumor microenvironment is addressed. Highlighted are examples of therapies used against each suppressive cell type, which vary from the hypothetical to the ineffective; the inefficient to the successful. A variety of treatments have been tried to combat this fundamental problem, indeed the cause that allows cancerous mutated cells to survive, multiply and overtake the body. Efficient methods to disable each particular suppressive type of cell have been introduced; this review summarizes the discussion with a table to guide future development. We see gene therapy as the most innovative and flexible method to lead the charge to specifically modifying the tumor microenvironment.
Collapse
Affiliation(s)
- Elizabeth A. Vasievich
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
50
|
Lee JC, Hayman E, Pegram HJ, Santos E, Heller G, Sadelain M, Brentjens R. In vivo inhibition of human CD19-targeted effector T cells by natural T regulatory cells in a xenotransplant murine model of B cell malignancy. Cancer Res 2011; 71:2871-81. [PMID: 21487038 DOI: 10.1158/0008-5472.can-10-0552] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human T cells genetically modified to express chimeric antigen receptors (CAR) specific to the B cell tumor antigen CD19 can successfully eradicate systemic human CD19(+) tumors in immunocompromised SCID (severe combined immunodeficient)-Beige mice. However, in the clinical setting, CD4(+) CD25(hi) T regulatory cells (Treg) present within the tumor microenvironment may be potent suppressors of tumor-targeted effector T cells. In order to assess the impact of Tregs on CAR-modified T cells in the SCID-Beige xenotransplant model, we isolated, genetically targeted and expanded natural T regulatory cells (nTreg). In vitro nTregs modified to express CD19-targeted CARs efficiently inhibited the proliferation of activated human T cells, as well as the capacity of CD19-targeted 19-28z(+) effector T cells to lyse CD19(+) Raji tumor cells. Intravenous infusion of CD19-targeted nTregs into SCID-Beige mice with systemic Raji tumors traffic to sites of tumor and recapitulate a clinically relevant hostile tumor microenvironment. Antitumor efficacy of subsequently infused 19-28z(+) effector T cells was fully abrogated as assessed by long-term survival of treated mice. Optimal suppression by genetically targeted nTregs was dependent on nTreg to effector T-cell ratios and in vivo nTreg activation. Prior infusion of cyclophosphamide in the setting of this nTreg-mediated hostile microenvironment was able to restore the antitumor activity of subsequently infused 19-28z(+) effector T cells through the eradication of tumor-targeted nTregs. These findings have significant implications for the design of future clinical trials utilizing CAR-based adoptive T-cell therapies of cancer.
Collapse
Affiliation(s)
- James C Lee
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|