Network


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

Hotspot


Dive into the research topics where Niloy J. Deb is active.

Publication


Featured researches published by Niloy J. Deb.


Oncology | 2006

Flt3L Therapy following Localized Tumor Irradiation Generates Long-Term Protective Immune Response in Metastatic Lung Cancer: Its Implication in Designing a Vaccination Strategy

Prabir K. Chakravarty; Chandan Guha; Alan A. Alfieri; Vivek Beri; Zoya Niazova; Niloy J. Deb; Zuoheng Fan; Elaine K. Thomas; Bhadrasain Vikram

Flt3 ligand (Flt3L) therapy that expands dendritic cells in vivo in combination with local tumor radiotherapy (RT) significantly improved survival and induced a long-term tumor-specific immune response in a murine model of Lewis lung carcinoma (3LL). The irradiated tumor cells were able to significantly restimulate the splenocytes of the RT + Flt3L cohort in vitro. The restimulated splenocytes demonstrated increased cytotoxic response, lymphocytic proliferation and elevated levels of Th type I cytokines (IL-2, IL-12, IFN-γ and TNF-α). The combination therapy of RT + Flt3L induced a long-term protective immunity in the disease-free animals. The protective effect was further enhanced when the disease-free animals were vaccinated with irradiated tumor cells. The vaccinated animals had significantly greater protection compared to the nonvaccinated group against subsequent challenge with 3LL cells. Taken together, these results indicate that the release of tumor antigens by irradiated dying tumors and concomitant administration of Flt3L was able to facilitate the generation of a tumor-specific long-term immune response against a poorly immunogenic tumor. This effect was further boosted by vaccination with irradiated tumor cells.


Gene Therapy | 2003

A novel strategy for in vivo expansion of transplanted hepatocytes using preparative hepatic irradiation and FasL-induced hepatocellular apoptosis

M Takahashi; Niloy J. Deb; Sung W. Lee; J Furgueil; T Okuyama; Namita Roy-Chowdhury; B Vikram; Jayanta Roy-Chowdhury; C Guha

A strategy for inducing preferential proliferation of the engrafted hepatocytes over host liver cells should markedly increase the benefit of hepatocyte transplantation for the treatment of liver diseases and ex vivo gene therapy. We hypothesized that preparative hepatic irradiation (HIR) to inhibit host hepatocellular regeneration in combination with the mitotic stimulus of host hepatocellular apoptosis should permit repopulation of the liver by transplanted cells. To test this hypothesis, congeneic normal rat hepatocytes were transplanted into UDP-glucuronosyltransferase (UGT1A1)-deficient jaundiced Gunn rats (a model of Crigler-Najjar syndrome type I), following HIR and adenovirus-mediated FasL gene transfer. Progressive repopulation of the liver by engrafted UGT1A1-proficient hepatocytes over 5 months was demonstrated by the appearance of UGT1A1 protein and enzyme activity in the liver, biliary bilirubin glucuronides secretion, and long-term normalization of serum bilirubin levels. This is the first demonstration of massive hepatic repopulation by transplanted cells by HIR and FasL-induced controlled apoptosis of host liver cells.


International Journal of Radiation Oncology Biology Physics | 2001

LIVER IRRADIATION: A POTENTIAL PREPARATIVE REGIMEN FOR HEPATOCYTE TRANSPLANTATION

Chandan Guha; Bhupesh Parashar; Niloy J. Deb; Anand Sharma; Giridhar R. Gorla; Alan A. Alfieri; Namita Roy-Chowdhury; Jayanta Roy-Chowdhury; Bhadrasain Vikram

Advances in the understanding of hepatocyte engraftment and repopulation of the host liver have already led to the use of hepatocyte transplantation (HT) with some success in the treatment of inherited and acquired liver diseases. Wider application of HT is severely limited by the unavailability of large number of transplantable hepatocytes and difficulties associated with transplanting an adequate number of cells for achieving therapeutically satisfactory levels of metabolic correction. Therefore, there is a need for preparative regimens that provide a growth advantage to the transplanted (healthy) hepatocytes over the hosts own (diseased) hepatocytes so that the former can repopulate the host liver. We have recently shown that when the liver of recipient rats was subjected to radiotherapy and partial hepatectomy before HT, the transplanted hepatocytes engrafted in and massively repopulated the liver, and also ameliorated the adverse clinical and histopathological changes associated with hepatic irradiation. This protocol was then used as a preparative regimen for transplanting normal hepatocytes into jaundice mutant rats (Gunn strain), which lack hepatic bilirubin-uridinediphosphoglucuronate glucuronosyltransferase and is a model of Crigler-Najjar syndrome Type I. The results showed long-term correction of the metabolic abnormality, suggesting that the transplanted hepatocytes repopulated an irradiated liver and were metabolically functional. This strategy could be useful in the treatment of various genetic, metabolic, or malignant diseases of the liver.


Radiation Research | 2014

An Autologous In Situ Tumor Vaccination Approach for Hepatocellular Carcinoma. 1. Flt3 Ligand Gene Transfer Increases Antitumor Effects of a Radio-Inducible Suicide Gene Therapy in an Ectopic Tumor Model

Niloy J. Deb; Madhur Garg; Rafi Kabarriti; Alan A. Alfieri; Masahiko Takahashi; Jayanta Roy-Chowdhury; Chandan Guha

Hepatocellular carcinoma (HCC) often presents as a diffuse or multifocal tumor making it difficult to control by surgery or radiation. Radio-inducible herpes simplex virus thymidine kinase (HSV-TK) gene therapy has been shown to enhance local tumor control after radiation therapy (RT), while limiting the expression of the transgene in the irradiated tumor tissues. To prevent liver tumor recurrence and control systemic disease while limiting the potential bystander toxicity of HSV-TK therapy, we proposed to stimulate endogenous dendritic cell (DC) proliferation with systemic adenovirus Flt3 ligand (Adeno-Flt3L) gene therapy, followed by primary tumor radiation therapy combined with a radio-inducible HSV-TK gene therapy. We hypothesized that adenovirus-expressing Flt3L gene therapy will stimulate DC proliferation, allowing the upregulated DCs to locally harness tumor antigens released from HSV-TK/RT-treated HCC cells, thereby converting irradiated tumors to an autologous in situ tumor vaccine in mice with primary liver tumors. To test this hypothesis, an expression vector of HSV-TK was constructed under the control of a radio-inducible promoter early-growth response (Egr-TK) and a recombinant adenovirus-expressing human Flt3L was constructed. The Adeno-Flt3L [109 plaque forming units (pfu)] was administered intravenously on days 1 and 8 after radiation therapy. The murine hepatoma cell line (BNL1ME) was stably transfected by Egr-TK or Egr-Null (encoding no therapeutic gene). Palpable tumors in BALB/c mice were treated with a localized dose of 25 Gy of radiation followed by ganciclovir (GCV, 100 mg/kg, 14 days). Four treatment cohorts were compared: Egr-Null/GCV + RT + Adeno-LacZ; Egr-Null/GCV + RT + Adeno-Flt3L; Egr-TK/GCV + RT + Adeno-LacZ; and Egr-TK/GCV + RT + Adeno-Flt3L. There was no primary tumor regression in the Egr-Null tumors after radiation therapy alone. In contrast, Egr-TK tumors had nearly complete tumor regression for 3 weeks after radiation therapy (P < 0.01), however, long-term follow-up demonstrated primary tumor recurrence and death secondary to pulmonary metastasis. Flt3L expression was confirmed by serum bioassay (mean = 88 ng/mL) in these animals and Western blotting of tissue culture medium in Adeno-Flt3L-infected BaF/huFlt3L cells. Radiation therapy with Adeno-Flt3L gene therapy effectively retarded primary tumor growth when compared to radiation therapy alone. The trimodality therapy (Egr-TK/GCV + RT + Adeno-Flt3L) was the most efficacious with 40% complete tumor regression (>100 days) and <20% pulmonary metastases, indicating the development of sustained antitumor immune response. These studies provide a rationale for triple modality therapies with radiation-inducible HSV-TK gene therapy and Adeno-Flt3L when used in combination with primary tumor radiation therapy for improved local and systemic control of HCC.


Radiation Research | 2014

An Autologous In Situ Tumor Vaccination Approach for Hepatocellular Carcinoma. 2. Tumor-Specific Immunity and Cure after Radio-Inducible Suicide Gene Therapy and Systemic CD40-Ligand and Flt3-Ligand Gene Therapy in an Orthotopic Tumor Model

Niloy J. Deb; Madhur Garg; Rafi Kabarriti; Zuoheng Fan; Alan A. Alfieri; Jayanta Roy-Chowdhury; Chandan Guha

Diffuse hepatocellular carcinoma (HCC) is a lethal disease that radiation therapy (RT) currently has a limited role in treating because of the potential for developing fatal radiation-induced liver disease. However, recently diffuse HCC, “radio-inducible suicide gene therapy” has been shown to enhance local tumor control and residual microscopic disease within the liver for diffuse HCC, by using a combination of chemoactivation and molecular radiosensitization. We have demonstrated that the addition of recombinant adenovirus-expressing human Flt3 ligand (Adeno-Flt3L) after radio-inducible suicide gene therapy induced a Th1-biased, immune response and enhanced tumor control in an ectopic model of HCC. We hypothesized that sequential administration of recombinant adenovirus-expressing CD40L (Adeno-CD40L) could further potentiate the efficacy of our trimodal therapy with RT + HSV-TK + Adeno-Flt3L. We examined our hypothesis in an orthotopic model of diffuse HCC using BNL1ME A.7R.1 (BNL) cells in Balb/c mice. BNL murine hepatoma cells (5 × 104) transfected with an expression vector of HSV-TK under the control of a radiation-inducible promoter were injected intraportally into BALB/cJ mice. Fourteen days after the HCC injection, mice were treated with a 25 Gy dose of radiation to the whole liver, followed by ganciclovir (GCV) treatment and systemic adenoviral cytokine gene therapy (Flt3L or CD40L or both). Untreated mice died in 27 ± 4 days. Radiation therapy alone had a marginal effect on survival (median = 35 ± 7 days) and the addition of HSV-TK/GCV gene therapy improved the median survival to 47 ± 6 days. However, the addition of Adeno-Flt3L to radiation therapy and HSV-TK/GCV therapy significantly (P = 0.0005) increased survival to a median of 63 ± 20 days with 44% (7/16) of the animals still alive 116 days after tumor implantation. The curative effect of Flt3L was completely abolished when using immunodeficient nude mice or mice depleted for CD4, CD8 and natural killer cells. The addition of Adeno-CD40L further improved the median survival of animals to 80 ± 15 days and this effect was abolished only when using anti-CD8 antibodies. Chromium-51 (51Cr) release assay showed cytotoxic T lymphocyte (CTL) activation, suggesting efficient dendritic cell (DC) activation with CTL activation after the treatment. Furthermore, when surviving mice were rechallenged with BNL-ETK cells on the foot pad, RT + HSV-TK/GCV + Flt3L + CD40L-treated mice developed a small tumor on day 56 but the tumor eventually disappeared after 105 days. Mice treated with RT + HSV-TK/GCV + Flt3L showed a slowed tumor growth curve compared with untreated mice. Therefore, combination therapy using Flt3L to induce DC proliferation and CD40L to enhance DC maturation holds great promise for immunomodulation of radiation therapy to enhance HCC tumor control and prevent progression of disease in patients with diffuse HCC.


Hepatology | 2002

Normal hepatocytes correct serum bilirubin after repopulation of Gunn rat liver subjected to irradiation/partial resection

Chandan Guha; Bhupesh Parashar; Niloy J. Deb; Madhur Garg; Giridhar R. Gorla; Anupam Singh; Namita Roy-Chowdhury; Bhadrasain Vikram; Jayanta Roy-Chowdhury


Artificial Organs | 2001

Amplification of Engrafted Hepatocytes by Preparative Manipulation of the Host Liver

Chandan Guha; Niloy J. Deb; Baljit S. Sappal; Siddhartha Sankar Ghosh; Namita Roy-Chowdhury; Jayanta Roy-Chowdhury


International Journal of Radiation Oncology Biology Physics | 2001

A novel therapeutic strategy for hepatocellular carcinoma: immunomodulation by Flt3-Ligand (Flt3L) following whole liver irradiation and radio-inducible HSV-TK gene therapy

Y. Kawashita; Niloy J. Deb; Madhur Garg; Z Fan; Alan A. Alfieri; S.J. Shah; P.K. Chakravarty; Jayanta Roy-Chowdhury; Bhadrasain Vikram; Chandan Guha


International Journal of Radiation Oncology Biology Physics | 2009

Treatment with R-Spondin 1 and Toll-like Receptor 9 Ligands Improve the Therapeutic Ratio of Abdominal Irradiation

Payel Bhanja; Subhrajit Saha; L. Liu; Rafi Kabarriti; H. Zhang; Niloy J. Deb; Madhur Garg; S. Kalnicki; Alan A. Alfieri; Chandan Guha


International Journal of Radiation Oncology Biology Physics | 2002

Radiation-enhanced in situ tumor vaccination for diffuse hepatocellular carcinoma: induction of tumor-specific immunity by systemic CD40-ligand and FLT3-Ligand gene therapy

Y. Kawashita; Niloy J. Deb; Madhur Garg; Z Fan; Alan A. Alfieri; S.J. Shah; P.K. Chakravarty; J Roy-Chowdhary; Bhadrasain Vikram; Chandan Guha

Collaboration


Dive into the Niloy J. Deb's collaboration.

Top Co-Authors

Avatar

Chandan Guha

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Alan A. Alfieri

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jayanta Roy-Chowdhury

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Madhur Garg

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Namita Roy-Chowdhury

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Bhupesh Parashar

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Giridhar R. Gorla

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rafi Kabarriti

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

L. Liu

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge