Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Front Immunol ; 14: 1245421, 2023.
Article En | MEDLINE | ID: mdl-38090567

Breast cancer (BC) is globally one of the leading killers among women. Within a breast tumor, a minor population of transformed cells accountable for drug resistance, survival, and metastasis is known as breast cancer stem cells (BCSCs). Several experimental lines of evidence have indicated that BCSCs influence the functionality of immune cells. They evade immune surveillance by altering the characteristics of immune cells and modulate the tumor landscape to an immune-suppressive type. They are proficient in switching from a quiescent phase (slowly cycling) to an actively proliferating phenotype with a high degree of plasticity. This review confers the relevance and impact of crosstalk between immune cells and BCSCs as a fate determinant for BC prognosis. It also focuses on current strategies for targeting these aberrant BCSCs that could open avenues for the treatment of breast carcinoma.


Breast Neoplasms , Mammary Neoplasms, Animal , Animals , Female , Humans , Breast Neoplasms/pathology , Mammary Neoplasms, Animal/pathology , Cell Line, Tumor , Prognosis , Neoplastic Stem Cells/metabolism
2.
Hum Immunol ; 83(5): 409-417, 2022 May.
Article En | MEDLINE | ID: mdl-35300874

In developing tumor, macrophages are one major immune infiltrate that not only contributes in shaping up of tumor microenvironment (TME) but also have the potential of determining the fate of tumor in terms of its progression. Phenotypic plasticity of macrophages primarily channelizes them to alternative (M2) form of tumor associated macrophages (TAM) in the TME. One of the key tumor derived components that plays a crucial role in TAM polarization from M1 to M2 form is lactic acid and has prominent role in progression of malignancy. The role of lactic acid as signalling molecule as well as an immunomodulator has recently been recognized. This review focuses on the mechanism and signalling that are involved in lactic acid induced M2 polarization and possible therapeutic strategies for regulating lactic acidosis in TME.


Neoplasms , Tumor Microenvironment , Humans , Lactic Acid , Macrophages , Tumor-Associated Macrophages
3.
Clin Immunol ; 232: 108875, 2021 11.
Article En | MEDLINE | ID: mdl-34740843

Inflammation is a part of carefully co-ordinated healing immune exercise to eliminate injurious stimuli. However, in substantial number of cancer types, it contributes in shaping up of robust tumor microenvironment (TME). Solid TME promotes infiltration of tumor associated macrophages (TAMs) that contributes to cancer promotion. TAMs are functionally heterogeneous and display an extraordinary degree of plasticity, which allow 'Switching' of macrophages into an 'M2', phenotype, linked with immunosuppression, advancement of tumor angiogenesis with metastatic consequences. In contrary to the classical M1 macrophages, these M2 TAMs are high-IL-10, TGF-ß secreting-'anti-inflammatory'. In this review, we will discuss the modes of infiltration and switching of TAMs into M2 anti-inflammatory state in the TME to promote immunosuppression and inflammation-driven cancer.


Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Animals , Humans
4.
Cell Immunol ; 316: 1-10, 2017 06.
Article En | MEDLINE | ID: mdl-28433198

Recent advances in tumor biology demand detailed analysis of the complex interaction of tumor cells with their adjacent microenvironment (tumor stroma) to understand the various mechanisms involved in tumor growth and metastasis. Mononuclear phagocytes or macrophages, a type of innate immune cells, defend the organism against infection and injury. On the otherhand, tumor associated macrophages (TAMs) constitute a significant part of the tumor-infiltrating immune cells, have been linked to the growth, angiogenesis, and metastasis of a variety of cancers, most likely through polarization of TAMs to the M2 (alternative) phenotype. Clinical and experimental evidences have shown that cancer tissues with high infiltration of TAMs are associated with poor patient prognosis and resistance to therapies, thus, targeting of TAMs in tumors is considered as a promising immunotherapeutic strategy. Depletion of M2 TAMs or 're-education' of them as anti-tumor effectors might contribute significantly to the search of new modalities in anti-cancer treatments. Basic questions on the factors responsible for homing of macrophages in tumors, mechanism of conversion of M1 to M2 TAMs, their functionality and, finally, the possible ways to target M2 TAMs are discussed.


Macrophages/immunology , Neoplasms/immunology , Tumor Microenvironment , Animals , Humans , Immunomodulation , Macrophages/pathology , Mice , Molecular Targeted Therapy , Neoplasms/therapy , Neovascularization, Pathologic , Tumor Escape
5.
Mol Immunol ; 80: 1-10, 2016 12.
Article En | MEDLINE | ID: mdl-27776244

Heterogeneous tumor microenvironment (TME), broadly divided into tumor core and peripheral sub-microenvironments, differentially polarize normal macrophages into a different form known as tumor associated M2 macrophages (M2TAMs) to promote tumor growth. In view of the extensive immune-editing role of NLGP, here, we have observed that NLGP is effective to convert M2TAMs (CD11b+F4/80high) to M1 (CD11b+F4/80low) more prominently in tumor core, along with downregulation of other M2 associated markers, like, ManR, Ym1, Fizz1. High IL-10:IL-12 ratio at tumor core was downregulated in NLGP treated melanoma bearing mice. Decrease in IL-10 by NLGP is again associated with the decrease in hypoxia, as indicated by prominent downregulation of HIF1α and VEGF, particularly at tumor core. Macrophages exposed to hypoxic tumor core lysates in vitro exhibited high IL-10, HIF1α and VEGF expression that was significantly downregulated by NLGP. Further evidences suggest M2TAM to M1 conversion by NLGP is associated with STAT3-regulated IL-10 dependent pathway without affecting the IL-4 dependent one. Such TAM modulatory functions of NLGP might help in the restriction of melanoma growth by increasing the proportion of M1 TAMs in tumor core that helps in prevention of tumor relapse and dissemination of the tumor mass.


Antineoplastic Agents, Phytogenic/pharmacology , Macrophages/immunology , Melanoma, Experimental/immunology , Plant Proteins/pharmacology , Signal Transduction/drug effects , Animals , Azadirachta , Blotting, Western , Cell Hypoxia/drug effects , Flow Cytometry , Glycoproteins/pharmacology , Immunohistochemistry , Interleukin-10/immunology , Macrophages/drug effects , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Plant Extracts/pharmacology , Plant Leaves , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
Immunol Lett ; 156(1-2): 7-17, 2013.
Article En | MEDLINE | ID: mdl-24029664

Content of tumor microenvironment (TME) is varied greatly among different types of laryngeal tumors, namely, supraglottic, glottic and subglottic tumors. These three different TMEs shape infiltrating monocytes/macrophages toward M2 genotypes in variable degrees. Results obtained from in vitro studies demonstrated extent of expression of M2 phenotypic features on macrophages was maximum after their exposure to supraglottic laryngeal tumor cell lysates (SLTCL) than glottic or subglottic lysates. Moreover, M2 macrophages generated under influence of SLTCL show less nitric oxide production, greater IL-10: IL-12 ratio and poor antigen presentation. Co-culture of such M2 macrophages with T cells from healthy donors resulted decreased activation of T cells and T cell mediated tumor cell cytotoxicity, than, glottic or subglottic. SLTCL mediated macrophage polarization is STAT3 dependent and might be one of the major factors for severe immune paralysis leading to poor prognosis of supraglottic laryngeal tumor bearer following standard treatment.


Immunologic Factors/metabolism , Laryngeal Neoplasms/metabolism , Macrophages/immunology , STAT3 Transcription Factor/immunology , Tissue Extracts/metabolism , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Epiglottis/metabolism , Epiglottis/pathology , Glottis/metabolism , Glottis/pathology , Humans , Immune Evasion/drug effects , Immune Evasion/immunology , Immunohistochemistry , Immunologic Factors/pharmacology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Laryngeal Neoplasms/pathology , Lymphocyte Activation/immunology , Macrophages/drug effects , Macrophages/metabolism , Male , Nitric Oxide/immunology , Nitric Oxide/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Tissue Extracts/pharmacology , Tumor Microenvironment
7.
J Immunol ; 191(2): 971-81, 2013 Jul 15.
Article En | MEDLINE | ID: mdl-23785117

Immune evasion within the tumor microenvironment supports malignant growth and is also a major obstacle for successful immunotherapy. Multiple cellular components and soluble factors coordinate to disrupt protective immune responses. Although stromal cells are well-known for their parenchymal supportive roles in cancer establishment and progression, we demonstrate for the first time, to our knowledge, that tumor-derived vascular pericytes negatively influence CD4(+) T cell activation and proliferation, and promote anergy in recall response to Ag by CD4(+)CD44(+) T cells via regulator of G protein signaling 5- and IL-6-dependent pathways. Our data support a new specific role for tumor-derived pericytes in the immune evasion paradigm within the tumor microenvironment and suggest the targeting of these cell populations in the context of successful immunotherapeutics for the treatment of cancer.


CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Neoplasms/immunology , Pericytes/metabolism , Tumor Escape , Animals , Bone Marrow Cells/immunology , Cell Line , Cell Proliferation , Chemokines/metabolism , Dendritic Cells/immunology , Female , GTP-Binding Proteins/metabolism , Histocompatibility Antigens Class II/metabolism , Hyaluronan Receptors/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , RGS Proteins/genetics , RGS Proteins/metabolism , RNA Interference , RNA, Small Interfering , Tumor Microenvironment/immunology
8.
PLoS One ; 8(6): e66501, 2013.
Article En | MEDLINE | ID: mdl-23785504

We have observed restriction of the murine sarcoma growth by therapeutic intervention of neem leaf glycoprotein (NLGP). In order to evaluate the mechanism of tumor growth restriction, here, we have analyzed tumor microenvironment (TME) from sarcoma bearing mice with NLGP therapy (NLGP-TME, in comparison to PBS-TME). Analysis of cytokine milieu within TME revealed IL-10, TGFß, IL-6 rich type 2 characters was switched to type 1 microenvironment with dominance of IFNγ secretion within NLGP-TME. Proportion of CD8(+) T cells was increased within NLGP-TME and these T cells were protected from TME-induced anergy by NLGP, as indicated by higher expression of pNFAT and inhibit related downstream signaling. Moreover, low expression of FasR(+) cells within CD8(+) T cell population denotes prevention from activation induced cell death. Using CFSE as a probe, better migration of T cells was noted within TME from NLGP treated mice than PBS cohort. CD8(+) T cells isolated from NLGP-TME exhibited greater cytotoxicity to sarcoma cells in vitro and these cells show higher expression of cytotoxicity related molecules, perforin and granzyme B. Adoptive transfer of NLGP-TME exposed T cells, but not PBS-TME exposed cells in mice, is able to significantly inhibit the growth of sarcoma in vivo. Such tumor growth inhibition by NLGP-TME exposed T cells was not observed when mice were depleted for CD8(+) T cells. Accumulated evidences strongly suggest NLGP mediated normalization of TME allows T cells to perform optimally to inhibit the tumor growth.


Azadirachta/chemistry , Glycoproteins/pharmacology , Plant Leaves/chemistry , Sarcoma/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects , Animals , Apoptosis/drug effects , Apoptosis/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Gene Expression Regulation, Neoplastic , Hypoxia , Lymphocyte Depletion , Mice , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Sarcoma/drug therapy , Sarcoma/genetics , Sarcoma/pathology , Tumor Microenvironment/immunology , fas Receptor/genetics , fas Receptor/immunology
9.
PLoS One ; 8(1): e47434, 2013.
Article En | MEDLINE | ID: mdl-23326300

In spite of sufficient data on Neem Leaf Glycoprotein (NLGP) as a prophylactic vaccine, little knowledge currently exists to support the use of NLGP as a therapeutic vaccine. Treatment of mice bearing established sarcomas with NLGP (25 µg/mice/week subcutaneously for 4 weeks) resulted in tumor regression or dormancy (Tumor free/Regressor, 13/24 (NLGP), 4/24 (PBS)). Evaluation of CD8(+) T cell status in blood, spleen, TDLN, VDLN and tumor revealed increase in cellular number. Elevated expression of CD69, CD44 and Ki67 on CD8(+) T cells revealed their state of activation and proliferation by NLGP. Depletion of CD8(+) T cells in mice at the time of NLGP treatment resulted in partial termination of tumor regression. An expansion of CXCR3(+) and CCR5(+) T cells was observed in the TDLN and tumor, along with their corresponding ligands. NLGP treatment enhances type 1 polarized T-bet expressing T cells with downregulation of GATA3. Treg cell population was almost unchanged. However, T∶Treg ratios significantly increased with NLGP. Enhanced secretion/expression of IFNγ was noted after NLGP therapy. In vitro culture of T cells with IL-2 and sarcoma antigen resulted in significant enhancement in cytotoxic efficacy. Consistently higher expression of CD107a was also observed in CD8(+) T cells from tumors. Reinoculation of sarcoma cells in tumor regressed NLGP-treated mice maintained tumor free status in majority. This is correlated with the increment of CD44(hi)CD62L(hi) central memory T cells. Collectively, these findings support a paradigm in which NLGP dynamically orchestrates the activation, expansion, and recruitment of CD8(+) T cells into established tumors to operate significant tumor cell lysis.


Antineoplastic Agents/immunology , CD8-Positive T-Lymphocytes/immunology , Glycoproteins/immunology , Plant Proteins/immunology , Sarcoma, Experimental/immunology , Animals , Antineoplastic Agents/pharmacology , Azadirachta/chemistry , Azadirachta/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Female , Glycoproteins/pharmacology , Immunohistochemistry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Plant Leaves/chemistry , Plant Leaves/immunology , Plant Proteins/pharmacology , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/pathology , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Survival Analysis , Time Factors , Tumor Burden/drug effects , Tumor Burden/immunology
10.
Int Immunopharmacol ; 12(2): 326-33, 2012 Feb.
Article En | MEDLINE | ID: mdl-22210373

We have shown that neem leaf glycoprotein (NLGP) inhibits the regulatory T cell (Tregs) induced suppression of tumoricidal functions of CD14(+)CD68(+) monocyte/macrophages (MO/Mφ) from human peripheral blood. Cytotoxic efficacy of MO/Mφ toward macrophage sensitive cells, U937, is decreased in presence of Tregs (induced), however, it was increased further by supplementation of NLGP in culture. Associated Treg mediated inhibition of perforin/granzyme B expression and nitric oxide release from MO/Mφ was normalized by NLGP. Altered status of signature cytokines, like, IL-12, IL-10, IL-6, TNFα from MO/Mφ under influence of Tregs is also rectified by NLGP. Tregs significantly enhanced the expression of altered marker, mannose receptor (CD206) on CD68(+) cells that was downregulated upon NLGP exposure. In addition to tumoricidal functions, antigen presenting ability of MO/Mφ is hampered by Treg induced downregulation of CD80, CD86 and HLA-ABC. NLGP upregulated these molecules in MO/Mφ even in the presence of Tregs. Treg mediated inhibition of MO/Mφ chemotaxis in contact dependent manner was also normalized partially by NLGP, where participation of CCR5 was documented. Overall results suggest that Treg influenced pro-tumor MO/Mφ functions are rectified in a significant extent by NLGP to create an anti-tumor immune environment.


Azadirachta/chemistry , Glycoproteins/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Plant Proteins/pharmacology , T-Lymphocytes, Regulatory/drug effects , Antigen Presentation/drug effects , Antigen Presentation/immunology , Azadirachta/immunology , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cytotoxicity, Immunologic/immunology , Glycoproteins/immunology , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Humans , Interleukins/immunology , Interleukins/metabolism , KB Cells , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Macrophages/immunology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Monocytes/immunology , Monocytes/metabolism , Nitric Oxide/immunology , Nitric Oxide/metabolism , Perforin/genetics , Perforin/immunology , Perforin/metabolism , Plant Leaves/chemistry , Plant Leaves/immunology , Plant Proteins/immunology , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
11.
Transl Res ; 158(4): 200-13, 2011 Oct.
Article En | MEDLINE | ID: mdl-21925117

Despite being a pleiotropic cytokine, the therapeutic potential of interferon α2b (IFNα2b) is debatable. Thus, the need for identifying predictive marker(s) for patients who are most likely to benefit from the treatment is pivotal for avoiding the exposure of nonresponsive patients to the toxicity of the treatment. To account for the attenuated efficacy of the drug, we have verified its dendritic cell (DC) maturating ability from monocytes of cervical cancer stage IIIB (CaCx-IIIB) patients. First, we evaluated the status of monocytes from CaCx-IIIB and healthy women by conducting flow cytometric studies of various activation markers and a cytokine analysis by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. Immature DCs were then generated from these monocytes and matured with low-dose IFNα2b (1500 units/mL). A functional and phenotypic comparative analysis of these matured DCs was performed by flow cytometric, proliferative, cytotoxic, and enzyme-linked immunosorbent assays. Our study shows that monocytes isolated from CaCx-IIIB are impaired, and in vitro maturation with IFNα2b did not significantly improve the functional repertoire of DCs generated from these monocytes in comparison with healthy controls. This impairment of monocytes might be a plausible reason for the attenuated efficacy of this drug alone in treating CaCx-IIIB patients, and this imbalance of immune parameters associated with the stage of malignancy might be considered an effective marker to design a proper therapeutic regimen.


Dendritic Cells/immunology , Interferon-alpha/adverse effects , Monocytes/immunology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/immunology , Adult , Aged , Antigen Presentation/drug effects , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/pathology , Female , Humans , In Vitro Techniques , Interferon alpha-2 , Interferon-alpha/therapeutic use , Lymphocyte Activation , Middle Aged , Monocytes/drug effects , Monocytes/pathology , Receptor, Interferon alpha-beta/metabolism , Recombinant Proteins , Translational Research, Biomedical , Treatment Failure , Uterine Cervical Neoplasms/pathology
12.
Immunotherapy ; 3(8): 949-69, 2011 Aug.
Article En | MEDLINE | ID: mdl-21843083

BACKGROUND: The presence of Tregs in tumors is associated with compromised tumor-specific immune responses and has a clear negative impact on survival of cancer patients. Thus, downregulation of Tregs is considered as a promising cancer immunotherapeutic approach. We have reported previously that neem leaf glycoprotein (NLGP) prophylaxis restricts tumor growth in mice by immune activation. In continuation, here, involvement of NLGP in the modulation of Tregs in association with tumor growth restriction is investigated. RESULTS: NLGP downregulates CD4+CD25+Foxp3+ Tregs within tumors. NLGP-mediated downregulation of CCR4 along with its ligand CCL22 restricts Treg migration at the tumor site. NLGP is not apoptotic to Tregs but significantly downregulates the expression of Foxp3, CTLA4 and GITR. It also reverses the functional impairment of T-effector cells by Tregs, in terms of IFN-γ secretion, cellular proliferation and tumor cell cytotoxicity. NLGP also facilitates reconditioning of tumor microenvironment (hostile) by increasing IFN-γ and IL-12 but decreasing IL-10, TGF-ß, VEGF and IDO, creating an antitumor niche. Interaction between Foxp3, p-NFATc3 and p-Smad2/3, needed for successful Treg function, is also inhibited by NLGP. CONCLUSION: All of these coordinated events might result in inhibition of Treg associated-tumor growth and therefore increased survivability of mice having NLGP treatment before or/and after tumor inoculation. Thus, the possibility of NLGP being an excellent tool as a T-cell anergy breaker by abrogating the suppressor functions of Tregs in cancer needs to be explored further in the clinic.


Antineoplastic Agents/administration & dosage , Glycoproteins/administration & dosage , Melanoma, Experimental/drug therapy , Plant Proteins/administration & dosage , Skin Neoplasms/drug therapy , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/adverse effects , Azadirachta , CD5 Antigens/biosynthesis , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Forkhead Transcription Factors/biosynthesis , Glycoproteins/adverse effects , Interleukin-2 Receptor alpha Subunit/biosynthesis , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Plant Leaves , Plant Proteins/adverse effects , Receptors, CCR4/genetics , Receptors, CCR4/metabolism , Signal Transduction/drug effects , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
...