Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kuntal Kanti Goswami is active.

Publication


Featured researches published by Kuntal Kanti Goswami.


PLOS ONE | 2013

Neem Leaf Glycoprotein Activates CD8 + T Cells to Promote Therapeutic Anti-Tumor Immunity Inhibiting the Growth of Mouse Sarcoma

Atanu Mallick; Subhasis Barik; Kuntal Kanti Goswami; Saptak Banerjee; Sarbari Ghosh; Koustav Sarkar; Anamika Bose; Rathindranath Baral

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 CD44hiCD62Lhi 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.


Journal of Immunology | 2013

Tumor-Derived Vascular Pericytes Anergize Th Cells

Anamika Bose; Subhasis Barik; Saptak Banerjee; Tithi Ghosh; Atanu Mallick; Suchandra Bhattacharyya Majumdar; Kuntal Kanti Goswami; Avishek Bhuniya; Sayantan Banerjee; Rathindranath Baral; Walter J. Storkus; Partha Sarathi Dasgupta; Subrata Majumdar

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.


PLOS ONE | 2013

Normalization of tumor microenvironment by neem leaf glycoprotein potentiates effector T cell functions and therapeutically intervenes in the growth of mouse sarcoma.

Subhasis Barik; Saptak Banerjee; Atanu Mallick; Kuntal Kanti Goswami; Soumyabrata Roy; Anamika Bose; Rathindranath Baral

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, TGFβ, 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.


Immunotherapy | 2011

Neem leaf glycoprotein inhibits CD4+CD25+Foxp3+ Tregs to restrict murine tumor growth.

Tathagata Chakraborty; Anamika Bose; Subhasis Barik; Kuntal Kanti Goswami; Saptak Banerjee; Shyamal Goswami; Diptendu Ghosh; Soumyabrata Roy; Krishnendu Chakraborty; Koustav Sarkar; Rathindranath Baral

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-β, 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.


Molecular Immunology | 2014

Targeting STAT3 phosphorylation by neem leaf glycoprotein prevents immune evasion exerted by supraglottic laryngeal tumor induced M2 macrophages

Kuntal Kanti Goswami; Subhasis Barik; Madhurima Sarkar; Anup Kumar Bhowmick; Jaydip Biswas; Anamika Bose; Rathindranath Baral

Tumor-associated macrophages (TAMs) are preferentially M2 skewed and promote tumor growth, angiogenesis, invasion, and/or metastasis. In this study, we have analyzed the in vitro immunomodulatory potential of a non-toxic neem leaf glycoprotein (NLGP) in reprogramming Stage III supraglottic laryngeal tumor cell lysate (SLTCL) induced M2 TAMs to their classical anti-tumor shape (M1). Data generated from this study support that NLGP is effective in preventing the SLTCL induced generation (CD68(+)CD206(+)IL-10(high) to CD68(+)CD206(-)IL-10(low) TAMs) and functions (NO(low) to NO(high), MHC-I(low) to MHC-I(high), CD80(low) to CD80(high)) of pro-tumorous M2 macrophages, which in turn associated with sustained anti-tumor effector functions by promoting cytotoxic T cell activities and suppressing regulatory T cells. Furthermore, our data also suggest that NLGP prevents M2 skewness of TAMs by downregulating phosphorylation of targeted STAT3.


International Immunopharmacology | 2013

Neem leaf glycoprotein is nontoxic to physiological functions of Swiss mice and Sprague Dawley rats: histological, biochemical and immunological perspectives.

Atanu Mallick; Sarbari Ghosh; Saptak Banerjee; Sayantani Majumder; Arnab Das; Bipasha Mondal; Subhasis Barik; Kuntal Kanti Goswami; Smarajit Pal; Subrata Laskar; Koustav Sarkar; Anamika Bose; Rathindranath Baral

We have evaluated the toxicity profile of a unique immunomodulator, neem leaf glycoprotein (NLGP) on different physiological systems of Swiss mice and Sprague Dawley rats. NLGP injection, even in higher doses than effective concentration caused no behavioral changes in animals and no death. NLGP injection increased the body weights of mice slightly without any change in organ weights. NLGP showed no adverse effect on the hematological system. Moreover, little hematostimulation was noticed, as evidenced by increased hemoglobin content, leukocyte count and lymphocyte numbers. Histological assessment of different organs revealed no alterations in the organ microstructure of the NLGP treated mice and rats. Histological normalcy of liver and kidney was further confirmed by the assessment of liver enzymes like alkaline phosphatase, SGOT, SGPT and nephrological products like urea and creatinine. NLGP has no apoptotic effect on immune cells but induces proliferation of mononuclear cells collected from mice and rats. Number of CD4(+), CD8(+) T cells, DX5(+) NK cells, CD11b(+) macrophages and CD11c(+) dendritic cells is upregulated by NLGP without a significant change in CD4(+)CD25(+)Foxp3(+) regulatory T cells. Type 1 cytokines, like IFNγ also increased in serum with a decrease in type 2 cytokines. Total IgG content, especially IgG2a increased in NLGP treated mice. These type 1 directed changes help to create an anti-tumor immune environment that results in the restriction of carcinoma growth in mice. Accumulated evidence strongly suggests the non-toxic nature of NLGP. Thus, it can be recommended for human use in anti-cancer therapy.


International Immunopharmacology | 2012

Neem leaf glycoprotein suppresses regulatory T cell mediated suppression of monocyte/macrophage functions.

Tathagata Chakraborty; Anamika Bose; Kuntal Kanti Goswami; Shyamal Goswami; Krishnendu Chakraborty; Rathindranath Baral

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.


Immunology Letters | 2013

Supraglottic laryngeal tumor microenvironmental factors facilitate STAT3 dependent pro-tumorigenic switch in tumor associated macrophages to render utmost immune evasion

Kuntal Kanti Goswami; Subhasis Barik; Saptak Banerjee; Anup Kumar Bhowmick; Jaydip Biswas; Anamika Bose; Rathindranath Baral

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.


Molecular Immunology | 2016

Neem leaf glycoprotein regulates function of tumor associated M2 macrophages in hypoxic tumor core: Critical role of IL-10/STAT3 signaling

Kuntal Kanti Goswami; Madhurima Sarkar; Sarbari Ghosh; Akata Saha; Tithi Ghosh; Ipsita Guha; Subhasis Barik; Saptak Banerjee; Soumyabrata Roy; Anamika Bose; Parthasarathi Dasgupta; Rathindranath Baral

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.


Human Immunology | 2012

Dysregulated CC receptor/ligand in monocytes/macrophages from tongue squamous cell carcinoma patients is partially rectified by interferon α-2b.

Krishnendu Chakraborty; Anamika Bose; Kuntal Kanti Goswami; Kalyan Kusum Mukherjee; Shyamal Goswami; Diptendu Ghosh; Tathagata Chakraborty; Koustav Sarkar; Smarajit Pal; Anup Kumar Bhowmick; Jaydip Biswas; Rathindranath Baral

In an aim to rectify dysregulated CC chemokine receptor (CCR5)/ligand (RANTES, MIP-1α, MIP-1β) status of monocytes/macrophages in tongue squamous cell carcinoma (TSCC; n = 12) patients, we have tested interferon α2b (IFNα2b), a novel immunomodulator with wide use in the management of several forms of cancer. IFNα2b can upregulate reduced CCR5 expression and increases the suppressed secretory status of its ligands, as evidenced from in vitro studies on monocytes/macrophages from the peripheral blood of TSCC patients as well as healthy individuals. Isolated monocytes of TSCC patients (n = 5) undergoing chemotherapeutic treatment along with IFNα2b immunotherapy demonstrated significant upregulation in CCR5 expression and secretion of corresponding ligands. These rectifications in receptor/ligand levels are reflected in improved CCR5-dependent migration of monocytes/macrophages after IFNα2b treatment. The rectified chemokine profile and cellular migration translate into better tumoricidal and antigen-presenting functions of these cells. Accordingly, enhanced T-cell-mediated tumor cell killing is demonstrated upon IFNα2b treatment. Translating dual benefits on monocyte/macrophage functions, IFNα2b may emerge as a potential form of immunotherapy for TSCC patients that may be combined with standard chemotherapy for better clinical outcome.

Collaboration


Dive into the Kuntal Kanti Goswami's collaboration.

Top Co-Authors

Avatar

Anamika Bose

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Rathindranath Baral

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Subhasis Barik

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Saptak Banerjee

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Koustav Sarkar

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Atanu Mallick

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jaydip Biswas

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Krishnendu Chakraborty

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Sarbari Ghosh

Chittaranjan National Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Shyamal Goswami

Chittaranjan National Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge