Network


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

Hotspot


Dive into the research topics where Debashish Bose is active.

Publication


Featured researches published by Debashish Bose.


Lancet Oncology | 2010

Vascular endothelial growth factor targeted therapy in the perioperative setting: implications for patient care

Debashish Bose; Funda Meric-Bernstam; Wayne L. Hofstetter; David A. Reardon; Keith T. Flaherty; Lee M. Ellis

Vascular endothelial growth factor (VEGF) targeted therapy, either alone or in combination with chemotherapy, has become the standard of care in several solid tumours, including colorectal cancer, renal-cell carcinoma, breast cancer, non-small-cell lung cancer, and glioblastoma. VEGF is crucial in the process of angiogenesis and wound healing and, thus, its inhibition has the potential to affect wound healing in patients undergoing surgery. In this review, we summarise the data available on the use of VEGF-targeted therapies, and their effect on perioperative wound complications. Surgery in patients receiving VEGF-targeted therapies seems to be safe when an appropriate interval of time is allowed between surgical procedures and treatment. Recommendations regarding this interval are provided in a disease and agent site-specific manner. We also discuss complications arising from the use of VEGF-directed therapies that might require surgical intervention and the considerations important in their management. At this juncture, safety data on the use of VEGF-targeted therapies in the perioperative period are sparse, and investigators are urged to continue to study this issue prospectively in current and future clinical trials to establish firm guidelines.


Clinical Cancer Research | 2009

Role of class 3 semaphorins and their receptors in tumor growth and angiogenesis.

Puja Gaur; Diane R. Bielenberg; Shaija Samuel; Debashish Bose; Yunfei Zhou; Michael J. Gray; Nikolaos A. Dallas; Fan Fan; Ling Xia; Jia Lu; Lee M. Ellis

Class 3 semaphorins (SEMA3) were first identified as glycoproteins that negatively mediate neuronal guidance by binding to neuropilin and repelling neurons away from the source of SEMA3. However, studies have shown that SEMA3s are also secreted by other cell types, including tumor cells, where they play an inhibitory role in tumor growth and angiogenesis (specifically SEMA3B and SEMA3F). SEMA3s primarily inhibit the cell motility and migration of tumor and endothelial cells by inducing collapse of the actin cytoskeleton via neuropilins and plexins. Besides binding to SEMA3s, neuropilin also binds the protumorigenic and proangiogenic ligand vascular endothelial growth factor (VEGF). Although some studies attribute the antitumorigenic and antiangiogenic properties of SEMA3s to competition between SEMA3s and VEGF for binding to neuropilin receptors, several others have shown that SEMA3s display growth-inhibitory activity independent of competition with VEGF. A better understanding of these molecular interactions and the role and signaling of SEMA3s in tumor biology will help determine whether SEMA3s represent potential therapeutic agents. Herein, we briefly review (a) the role of SEMA3s in mediating tumor growth, (b) the SEMA3 receptors neuropilins and plexins, and (c) the potential competition between SEMA3s and VEGF family members for neuropilin binding. (Clin Cancer Res 2009;15(22):676370)


British Journal of Cancer | 2011

Chronic exposure of colorectal cancer cells to bevacizumab promotes compensatory pathways that mediate tumour cell migration.

Fan Fan; Shaija Samuel; Puja Gaur; Jia Lu; Nikolaos A. Dallas; Ling Xia; Debashish Bose; Lee M. Ellis

Background:Bevacizumab (Bev), a monoclonal antibody to vascular endothelial growth factor (VEGF), is used in combination with chemotherapy for the treatment of metastatic colorectal cancer (CRC). The effects of Bev on angiogenesis have been well described, but the direct effect of Bev on tumour cells is unknown. This study was carried out to determine the molecular and phenotypic changes in CRC cells after chronic Bev exposure in vitro.Methods:Human CRC cell lines were chronically exposed (3 months) to Bev in vitro to develop Bev-adapted (Bev-A) cell lines. Vascular endothelial growth factor family members were determined by reverse transcription–polymerase chain reaction and western blotting. Migration and invasion was determined using standard in vitro assays. Intravenous injection of tumour cells was carried out to evaluate metastatic potential in mice.Results:Bevacizumab-adapted cells were found to be more migratory and invasive than control cells (P<0.001). Bevacizumab-adapted cells showed higher levels of VEGF-A, -B, -C, placental growth factor (PlGF), VEGF receptor-1 (VEGFR-1) and phosphorylation of VEGFR-1. Furthermore, treatment with SU5416, a VEGFR protein tyrosine kinase inhibitor, led to significantly decreased cell migration in vitro (P<0.001). Bevacizumab-adapted cells were more metastatic in vivo (P<0.05).Conclusion:Chronic exposure of CRC cells to Bev (1) increased expression of VEGF-A, -B, -C, PlGF, VEGFR-1 and VEGFR-1 phosphorylation, (2) increased tumour cell migration and invasion, and (3) metastatic potential in vivo. Our study shows the functional significance of autocrine VEGF signalling in CRC cells.


British Journal of Cancer | 2011

Chemoresistant colorectal cancer cells and cancer stem cells mediate growth and survival of bystander cells

Debashish Bose; L. J. Zimmerman; Mariaelena Pierobon; Emmanuel Petricoin; Federico Tozzi; Alexander A. Parikh; Fan Fan; Nikolaos A. Dallas; Ling Xia; Puja Gaur; Shaija Samuel; Daniel C. Liebler; Lee M. Ellis

Background:Recent studies suggest that cancer stem cells (CSCs) mediate chemoresistance, but interestingly, only a small percentage of cells in a resistant tumour are CSCs; this suggests that non-CSCs survive by other means. We hypothesised that chemoresistant colorectal cancer (CRC) cells generate soluble factors that enhance survival of chemonaive tumour cells.Methods:Chemoresistant CRC cells were generated by serial passage in oxaliplatin (Ox cells). Conditioned media (CM) was collected from parental and oxaliplatin-resistant (OxR) cells. CRC cells were treated with CM and growth and survival were assessed. Tumour growth rates were determined in nude mice after cells were treated with CM. Mass spectrometry (MS) identified proteins in CM. Reverse phase protein microarray assays determined signalling effects of CM in parental cells.Results:Oxaliplatin-resistant CM increased survival of chemo-naive cells. CSC CM also increased growth of parental cells. Parental and OxR mixed tumours grew larger than tumours composed of parental or OxR cells alone. Mass spectrometry detected unique survival-promoting factors in OxR CM compared with parental CM. Cells treated with OxR CM demonstrated early phosphorylation of EGFR and MEK1, with later upregulation of total Akt .We identified progranulin as a potential mediator of chemoresistance.Conclusion:Chemoresistant tumour cells and CSCs may promote resistance through soluble factors that mediate survival in otherwise chemosensitive tumour cells.


Seminars in Oncology | 2009

Targeting tumor angiogenesis.

Puja Gaur; Debashish Bose; Shaija Samuel; Lee M. Ellis

Our understanding of the process of tumor angiogenesis has changed significantly since the late 1970s, when vascular endothelial growth factor (VEGF) was first identified as vascular permeability factor and later found to be the major mediator of physiologic and pathologic angiogenesis. Since then, several additional VEGF-related ligands, VEGF receptors (VEGFRs), and complementary/alternative pathways that regulate tumor angiogenesis have been identified. Over the last decade, several antiangiogenic agents have been developed with the aim to inhibit new blood vessel growth, and we have learned that VEGF inhibition does far more than simply block new blood vessel growth. Clinical studies have demonstrated an improvement of progression-free and overall survivals with anti-VEGF therapy (with or without chemotherapy) in patients with advanced-stage malignancies. Unfortunately, even when anti-VEGF therapy is effective, the benefit of therapy is short-lived, with the development of tumor growth. We now recognize the presence of numerous complementary and redundant pathways that regulate tumor vasculature. For example, VEGF/VEGFR and angiopoietin/Tie-2 axes are two redundant, complementary components regulating tumor angiogenesis and vascular maintenance. The current clinical challenge is to identify: (1) factors that predict efficacy, and (2) markers of tumor response to anti-VEGF therapy, which can be achieved only by developing a thorough understanding of the biology of the VEGF system and the role of complementary pathways that may mediate resistance to anti-VEGF therapy.


Journal of The American College of Surgeons | 2010

Multidisciplinary management strategy for incidental cystic lesions of the pancreas.

Debashish Bose; Eric P. Tamm; Jun Liu; Leonardo P. Marcal; Aparna Balachandran; Priya Bhosale; Jason B. Fleming; Jeffrey E. Lee; Douglas B. Evans; Rosa F. Hwang

BACKGROUND At our institution, incidental pancreatic cysts are frequently identified in asymptomatic patients undergoing routine imaging for staging of nonpancreatic malignancies. Management of these patients is unclear because a small but significant number of incidental pancreatic cysts are malignant. STUDY DESIGN Our institutional database was reviewed for patients with ICD-9 codes for pancreatic cysts from 1980 to 2005. Clinicopathologic factors, including CT and endoscopic ultrasound (EUS) characteristics and management strategies, were analyzed. RESULTS Over 25 years, 942 patients were identified with pancreatic cysts. Excluding those with symptoms or pseudocysts, 350 patients remained with incidental pancreatic cysts. Mean overall survival was 41.4 months (mean follow-up 32.7 months). Forty-one patients underwent resection, of whom 38 (92.7%) had premalignant or malignant pathology. Univariate analysis of variables predicting pathologic premalignant or malignant diagnosis identified pancreatic neck or body location as significant factors. CONCLUSIONS These data suggest that most incidental pancreatic cysts can be managed nonoperatively using a selective strategy based on detailed review of CT imaging and EUS findings.


PLOS ONE | 2011

Neuropilin-2 mediated β-catenin signaling and survival in human gastro-intestinal cancer cell lines

Shaija Samuel; Puja Gaur; Fan Fan; Ling Xia; Michael J. Gray; Nikolaos A. Dallas; Debashish Bose; Cristian Rodriguez-Aguayo; Gabriel Lopez-Berestein; Greg Plowman; Anil Bagri; Anil K. Sood; Lee M. Ellis

NRP-2 is a high-affinity kinase-deficient receptor for ligands belonging to the class 3 semaphorin and vascular endothelial growth factor families. NRP-2 has been detected on the surface of several types of human cancer cells, but its expression and function in gastrointestinal (GI) cancer cells remains to be determined. We sought to determine the function of NRP-2 in mediating downstream signals regulating the growth and survival of human gastrointestinal cancer cells. In human gastric cancer specimens, NRP-2 expression was detected in tumor tissues but not in adjacent normal mucosa. In CNDT 2.5 cells, shRNA mediated knockdown NRP-2 expression led to decreased migration and invasion in vitro (p<0.01). Focused gene-array analysis demonstrated that loss of NRP-2 reduced the expression of a critical metastasis mediator gene, S100A4. Steady-state levels and function of β-catenin, a known regulator of S100A4, were also decreased in the shNRP-2 clones. Furthermore, knockdown of NRP-2 sensitized CNDT 2.5 cells in vitro to 5FU toxicity. This effect was associated with activation of caspases 3 and 7, cleavage of PARP, and downregulation of Bcl-2. In vivo growth of CNDT 2.5 cells in the livers of nude mice was significantly decreased in the shNRP-2 group (p<0.05). Intraperitoneal administration of NRP-2 siRNA-DOPC decreased the tumor burden in mice (p = 0.01). Collectively, our results demonstrate that tumor cell–derived NRP-2 mediates critical survival signaling in gastrointestinal cancer cells.


Cancer Research | 2011

Abstract 4075: Chemoresistant colorectal cancer cells exhibit high glycolytic activity

Federico Tozzi; Yunfei Zhou; Jinyu Chen; Debashish Bose; Fan Fan; Ling Xia; Jinrong Wang; Heather Brusher; William R. Widger; Lee M. Ellis; Zhang Waihua

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Background: Overcoming drug resistance in colorectal cancer (CRC) requires an understanding the mechanisms by which cancer cells adapt to the genotoxic stress. We established oxaliplatin resistant CRC cells derived from HCT116 and HT29 cell lines (HCT116 OxR and HT29 OxR) (Yang AD et al, Clin Ca Res, 2007). The mechanisms by which these resistant clones reprogram their energy metabolism to gain a survival advantage remain unknown. In this study, we hypothesized that chemoresistant CRC cells would exhibit altered cellular metabolism in order to survive following chronic genotoxic stress. Methods: The expression level of the glycolytic enzymes Glut1, hexokinase II (HK2), LDHA and HIF1α were detected by Western Blotting. Glucose uptake and lactate production was calculated by measuring the concentration of glucose and lactate in the culture media. Intracellular ATP/ADP levels, oxygen consumption and mitochondria ATP production were quantified. An in vivo xenograft study was used to compare the growth rate and angiogenesis of parental and oxaliplatin-resistant cells. Results: Compared with the parental cells, both HT29-OxR and HCT116-OxR cells exhibited a metabolic phenotype with increased glycolysis as reflected by increased glucose uptake and lactate production. Glycolytic enzymes were upregulated in resistant cells including Glut1, HK2 and LDHA. HIF1α expression and VEGF levels in the conditioned media were increased in the resistant cells. The mitochondria of OxR cells demonstrated defective complex I/II substrate ATP production despite increased cellular oxygen consumption. Importantly, the OxR cells maintain higher levels of intracellular ATP and ATP/ADP ratio indicating a metabolic switch to glycolysis. In a tumor xenograft model, HT29 OxR cells grew significantly slower than parental HT29 cells. Interestingly, when OxR cells were mixed with parental HT29 cells (50:50 and 90:10), tumor growth and microvessel counts were significantly increased. Conclusion: Oxaliplatin-resistant cells demonstrated: 1) high aerobic glycolytic activity, 2) an increase in HIF1α and glycolytic enzymes, and 3) defective mitochondria function. This metabolic switch provides more ATP production and likely contributes to the chemoresistant phenotype. Altering energy metabolism may provide a novel strategy to overcome drug-resistance in CRC cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4075. doi:10.1158/1538-7445.AM2011-4075


Cancer Research | 2010

Abstract 2276: Proteomic analysis of chemoresistance in colorectal cancer cells: potential paracrine mechanisms of resistance

Debashish Bose; Fan Fan; Federico Tozzi; Lisa J. Zimmerman; Matthew H. Herynk; Mariaelena Pierobon; Shaija Samuel; Robbert J.C. Slebos; Alexander A. Parikh; Emmanuel Petricoin; Daniel C. Liebler; Lee M. Ellis

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Background: Chemoresistance occurs in nearly all patients with metastatic colorectal cancer (CRC), and mechanisms to reverse chemoresistance remain elusive. We tested the hypothesis that CRC cells resistant to 5-fluorouracil (5FU-R) and oxaliplatin (Ox-R) exhibit proteomic profiles that may identify previously unrecognized mediators of resistance. In prior studies, we found that conditioned media from oxaliplatin-resistant HT29 cells (Ox-R) could mediate growth and chemoresistance in chemonaive parental HT29 cells in vitro. We sought to identify soluble factors in conditioned media that are potential mediators of the paracrine cell survival mechanisms. Methods: Parental HT29 (Par) cells were grown in increasing concentrations of 5-FU and oxaliplatin to generate 5FU-R and Ox-R cells. Protein from cell lysates and conditioned media (CM) were analyzed by liquid chromatography-mass spectrometry (LC-MS), and spectral counts were compared. Antibody-conjugated bead technology and ELISAs were used to obtain cytokine profiles of CM. Reverse phase proteomic arrays (RPMA) were used to determine signal transduction pathways activated in cells treated with CM. Ox-R cells were injected into nude mice to determine the paracrine effect on Par cells growing on the opposite flank. Results: Chemoresistant cells displayed significantly different proteomic profiles. In 5FU-R cells, pathways involving oxidative phosphorylation, inositol metabolism, actin cytoskeleton signaling, regulation of actin-based motility by Rho, and ATM signaling were significantly altered vs Par cells. In Ox-R cells, pathways mediating pyruvate metabolism, integrin signaling, caveolar-mediated endocytosis signaling, and mitochondrial dysfunction were among the most altered vs Par cells. Comparison of 5FU-R and Ox-R cells revealed differences in RNA post-transcriptional modification, ERK/MAPK, RAN, and chemokine signaling. Cytokine profiling demonstrated a significant increase in stem cell factor/c-Kit ligand (p=0.02) and a decrease in TRAIL (p=0.008) and IL-10 (p=0.04). RPPA analysis demonstrated early phosphorylation of EGFR and MEK1 followed by GSK, and mTOR activation. Ox-R tumors growing in vivo induced faster and larger tumor growth of contralateral Par tumors indicating a systemic effect. Conclusions: Chemoresistant CRC cells exhibit proteomes that reflect specific survival pathways, with many previously unrecognized potential mediators of resistance. Analysis of soluble factors from chemoresistant CRC cells demonstrates the presence of numerous potential mediators of cancer cell survival that may act, not only in an autocrine/paracrine manner, but also systemically. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2276.


Cancer Research | 2010

Abstract 1281: Endothelial cells promote the cancer stem cell phenotype of human colorectal cancer cells

Jia Lu; Debashish Bose; Yunfei Zhou; Puja Gaur; Shaija Samuel; Fan Fan; Ling Xia; Xiangcang Ye; Eric Sceusi; Federico Tozzi; Sendurai A. Mani; Lee M. Ellis

Background: Cancer stem cells (CSC), which possess the properties of self-renewal, multi-potential differentiation and tumor initiation, can arise either from normal stem cells or due to the influence of the tumor micro-environment. This study was done to determine the potential role of endothelial cell (EC) derived paracrine factors on promoting the CSC phenotype in human colorectal cancer (CRC) cells. Methods: RFP-labeled human CRC cells HCT116 were co-cultured with RF24 ECs (immortalized HUVECs) under low serum conditions. After FAC-sorting for the CRC cells, the CSC population was analyzed by 1) the Aldefluor assay, 2) sphere forming ability, and 3) Western blot analysis for the CSC markers CD133 and CD44. In addition, low serum condition media obtained from either ECs or parental CRC cells (control), was added to CRC cells to determine its effect on the CSC phenotype. In order to characterize the apoptotic effect of EC derived factors on CRC cells, we performed Annexin V staining by FACS, PARP cleavage, Caspase 3 cleavage, and Bcl2 levels by Western blotting. Using MTT assay, we determined the effect of EC derived factors on chemo-sensitivity of CRC cells exposed to 5-FU. Results: Co-culturing of CRC cells with ECs markedly increased the ALDH-positive population from 5% to 18%, and the sphere forming ability by more than 4-fold in CRC cells. In addition, CRC cells also displayed increased CD133 (6-fold) and CD44 (4-fold) protein levels. Furthermore, this effect could be mimicked simply by co-culturing CRC cells with conditioned media obtained from ECs. Similarly, treatment of CRC cells with conditioned medium from ECs significantly increased the ALDH-positive population from 4% to 8%, sphere forming ability >3-fold, and the expression of CD133 (7-fold) and CD44 (6-fold). Conditioned medium from ECs, concomitantly decreased spontaneous apoptosis in CRC cells as demonstrated by a 3-fold decrease in Annexin V-positive population (21% to 7%), down-regulation of cleaved PARP and Caspase 3, and up-regulation of Bcl2. CRC cells exposed to EC conditioned medium also displayed decreased sensitivity to 5-FU [>10-fold increase of the IC50 from 2.0 μg/ml to 22.3 μg/ml (p Conclusions: Exposure of CRCs to ECs increased CSC properties, and decreased spontaneous apoptosis. Since, this effect can also be mimicked by the conditioned media, additional experiments to identify and validate the soluble factors that mediate this effect are in progress. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1281.

Collaboration


Dive into the Debashish Bose's collaboration.

Top Co-Authors

Avatar

Lee M. Ellis

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Fan Fan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shaija Samuel

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ling Xia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Puja Gaur

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Federico Tozzi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nikolaos A. Dallas

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jia Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yunfei Zhou

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge