57 results on '"Mallya K"'
Search Results
2. A rare and interesting case of double intussusception
- Author
-
Mallya, K. Ashwin, Sandhya, Y.S., Salil, Rajayer, Neeraj, Chaudhri, and Nayak, B.M.
- Subjects
Intestines -- Intussusception ,Intestines -- Case studies ,Intestines -- Diagnosis ,Health - Abstract
Intussusception is a rare cause of intestinal obstruction in adults. Ninety percent of these patients have a pathologic lead point. Here we present a case of double intussusception in a 46-year-old lady. She presented with features of acute intestinal obstruction with a preceding history of recurrent constipation, mass per rectum on defecation and severe mucus discharge per rectum. Laparotomy revealed a double intussusception with a prograde ileo-ceco-colocolic intusussception due to a submucous lipoma in the caecum and a retrograde sigmoido-descending intussusception which revealed itself after reducing the prograde intussusception. Purely retrograde intussusceptions are extremely rare and only 26 such cases have been reported so far in the literature. Retrograde intussusception with an antegrade component due to a submucous lipoma which had completed its course into the rectum has not been reported so far., Table of Contents Abstract Introduction Case Report Discussion Conclusion References Introduction Intussusception is an invagination of one segment (intussusceptum) of intestine into another (intussuscipiens). Gross (1) series of 102 cases [...]
- Published
- 2009
3. Tuberculosis presenting as a ruptured aneurysm of the internal carotid artery
- Author
-
Ashwin, Mallya K., Salil, Rajayer, Sandhya, Y.S., Ashok, Shetty, Madhav, Kamath, and Prem, Prakash
- Subjects
Aortic aneurysms -- Risk factors ,Aortic aneurysms -- Diagnosis ,Aortic aneurysms -- Care and treatment ,Aortic aneurysms -- Patient outcomes ,CT imaging -- Usage ,CT imaging -- Health aspects ,Tuberculosis -- Risk factors ,Tuberculosis -- Diagnosis ,Tuberculosis -- Drug therapy ,Tuberculosis -- Patient outcomes ,Tuberculosis -- Case studies ,Health - Abstract
Tuberculosis is often included in the differential diagnosis for almost every set of symptoms that a patient may present to the hospital with. We report a case of extrapulmonary tuberculosis presenting as a life threatening emergency. A 51 year old male presented with a swelling on the right side of the neck and a sinus that was discharging serosanguinous fluid. A Doppler study revealed a false aneurysm of the internal carotid artery which ruptured before any elective management could be instituted. He was resuscitated and subjected to an emergency CT angiogram which confirmed the diagnosis. This was followed by right internal carotid artery ligation and an excision biopsy of lymph nodes in the field which revealed on histopathology to be of tuberculous origin. He was a known case of pulmonary tuberculosis who had defaulted on treatment. This case is highlighted to stress upon an uncommon manifestation of a very common and non malignant disease. Keywords: Tuberculosis, Aneurysm, Carotid, Lymphadenopathy, Table of Contents Abstract Introduction Case Report Discussion Conclusion References Introduction A false aneurysm (or pseudoaneurysm) is a breach in the vascular wall leading to an extravascular haematoma that freely [...]
- Published
- 2008
4. Secure learning in the mobile cloud
- Author
-
Mallya, K Ramananda, primary and Dhas, C Suresh Gnana, additional
- Published
- 2016
- Full Text
- View/download PDF
5. Potentials of Plasma NGAL and MIC-1 as Biomarker(s) in the Diagnosis of Lethal Pancreatic Cancer
- Author
-
Kaur, S, Chakraborty, S, Baine, MJ, Mallya, K, Smith, LM, Sasson, A, Brand, R, Guha, S, Jain, M, Wittel, U, Singh, SK, Batra, SK, Kaur, S, Chakraborty, S, Baine, MJ, Mallya, K, Smith, LM, Sasson, A, Brand, R, Guha, S, Jain, M, Wittel, U, Singh, SK, and Batra, SK
- Abstract
Pancreatic cancer (PC) is lethal malignancy with very high mortality rate. Absence of sensitive and specific marker(s) is one of the major factors for poor prognosis of PC patients. In pilot studies using small set of patients, secreted acute phase proteins neutrophil gelatinase associated lipocalin (NGAL) and TGF-β family member macrophage inhibitory cytokine-1 (MIC-1) are proposed as most potential biomarkers specifically elevated in the blood of PC patients. However, their performance as diagnostic markers for PC, particularly in pre-treatment patients, remains unknown. In order to evaluate the diagnostic efficacy of NGAL and MIC-1, their levels were measured in plasma samples from patients with pre-treatment PC patients (n = 91) and compared it with those in healthy control (HC) individuals (n = 24) and patients with chronic pancreatitis (CP, n = 23). The diagnostic performance of these two proteins was further compared with that of CA19-9, a tumor marker commonly used to follow PC progression. The levels of all three biomarkers were significantly higher in PC compared to HCs. The mean (± standard deviation, SD) plasma NGAL, CA19-9 and MIC-1 levels in PC patients was 111.1 ng/mL (2.2), 219.2 U/mL (7.8) and 4.5 ng/mL (4.1), respectively. In comparing resectable PC to healthy patients, all three biomarkers were found to have comparable sensitivities (between 64%-81%) but CA19-9 and NGAL had a higher specificity (92% and 88%, respectively). For distinguishing resectable PC from CP patients, CA19-9 and MIC-1 were most specific (74% and 78% respectively). CA19-9 at an optimal cut-off of 54.1 U/ml is highly specific in differentiating resectable (stage 1/2) pancreatic cancer patients from controls in comparison to its clinical cut-off (37.1 U/ml). Notably, the addition of MIC-1 to CA19-9 significantly improved the ability to distinguish resectable PC cases from CP (p = 0.029). Overall, MIC-1 in combination with CA19-9 improved the diagnostic accuracy of differentiatin
- Published
- 2013
6. Transcriptional profiling of peripheral blood mononuclear cells in pancreatic cancer patients identifies novel genes with potential diagnostic utility
- Author
-
Baine, MJ, Chakraborty, S, Smith, LM, Mallya, K, Sasson, AR, Brand, RE, Batra, SK, Baine, MJ, Chakraborty, S, Smith, LM, Mallya, K, Sasson, AR, Brand, RE, and Batra, SK
- Abstract
Background: It is well known that many malignancies, including pancreatic cancer (PC), possess the ability to evade the immune system by indirectly downregulating the mononuclear cell machinery necessary to launch an effective immune response. This knowledge, in conjunction with the fact that the trancriptome of peripheral blood mononuclear cells has been shown to be altered in the context of many diseases, including renal cell carcinoma, lead us to study if any such alteration in gene expression exists in PC as it may have diagnostic utility. Methods and Findings: PBMC samples from 26 PC patients and 33 matched healthy controls were analyzed by whole genome cDNA microarray. Three hundred eighty-three genes were found to be significantly different between PC and healthy controls, with 65 having at least a 1.5 fold change in expression. Pathway analysis revealed that many of these genes fell into pathways responsible for hematopoietic differentiation, cytokine signaling, and natural killer (NK) cell and CD8+ T-cell cytotoxic response. Unsupervised hierarchical clustering analysis identified an eight-gene predictor set, consisting of SSBP2, Ube2b-rs1, CA5B, F5, TBC1D8, ANXA3, ARG1, and ADAMTS20, that could distinguish PC patients from healthy controls with an accuracy of 79% in a blinded subset of samples from treatment naïve patients, giving a sensitivity of 83% and a specificity of 75%. Conclusions: In summary, we report the first in-depth comparison of global gene expression profiles of PBMCs between PC patients and healthy controls. We have also identified a gene predictor set that can potentially be developed further for use in diagnostic algorithms in PC. Future directions of this research should include analysis of PBMC expression profiles in patients with chronic pancreatitis as well as increasing the number of early-stage patients to assess the utility of PBMCs in the early diagnosis of PC. © 2011 Baine et al.
- Published
- 2011
7. Cerebrovasculr response to CO2 in edematous brain during either fentanyl or isoflurane anesthesia.
- Author
-
Shah, Nitin, Long, Charles, Marx, William, DiResta, Gene R., Arbit, Ehund, Mascott, Christopher, Mallya, Kasargod, Bedford, Robert, Shah, N, Long, C, Marx, W, DiResta, G R, Arbit, E, Mascott, C, Mallya, K, and Bedford, R
- Published
- 1990
- Full Text
- View/download PDF
8. CEREBROVASCULAR RESPONSE TO CO, IN EDEMATOUS BRAIN DURING EITHER FENTANYL OR ISOFLURANE ANESTHESIA
- Author
-
SHAH, N. K., primary, LONG, C. W., additional, MARX, W., additional, DiRESTA, G. R., additional, ARBIT, E., additional, MASCOTT, C., additional, MALLYA, K., additional, and BEDFORD, R. F., additional
- Published
- 1988
- Full Text
- View/download PDF
9. Targeting Human Pancreatic Cancer with a Fluorophore-Conjugated Mucin 4 (MUC4) Antibody: Initial Characterization in Orthotopic Cell Line Mouse Models.
- Author
-
Jaiswal S, Cox KE, Amirfakhri S, Din Parast Saleh A, Kobayashi K, Lwin TM, Talib S, Aithal A, Mallya K, Jain M, Mohs AM, Hoffman RM, Batra SK, and Bouvet M
- Abstract
Background/Objectives: Pancreatic cancer is the third leading cause of death related to cancer. The only possible cure presently is complete surgical resection; however, this is limited by difficulty in clearly defining tumor margins. Enhancement of the visualization of pancreatic ductal adenocarcinoma (PDAC) tumor margins using near-infrared dye-conjugated tumor-specific antibodies was pioneered by using anti-CEA, anti-CA19.9, and anti-MUC5AC in orthotopic mouse models of pancreatic cancer. Recently, an antibody to Mucin 4 (MUC4) conjugated to a fluorescent probe has shown promise in targeting colon tumors in orthotopic mouse models. Methods: In the present study, we targeted pancreatic cancer using an anti-MUC4 antibody conjugated to IRDye800 (anti-MUC4-IR800) in orthotopic mouse models. Two pancreatic cancer human cell lines were used, SW1990 and CD18/HPAF. Results: Anti-MUC4-IR800 targeted the two pancreatic cancer cell line tumors in orthotopic mouse models with high tumor-to-pancreas ratios and high tumor-to-liver ratios, with greater targeting seen in SW1990. Conclusions: The present results suggest anti-MUC4-IR800's potential to be used in fluorescence-guided surgical resection of pancreatic cancer.
- Published
- 2024
- Full Text
- View/download PDF
10. Chimeric antibody targeting unique epitope on onco-mucin16 reduces tumor burden in pancreatic and lung malignancies.
- Author
-
Shah A, Chaudhary S, Lakshmanan I, Aithal A, Kisling SG, Sorrell C, Marimuthu S, Gautam SK, Rauth S, Kshirsagar P, Cox JL, Natarajan G, Bhatia R, Mallya K, Rachagani S, Nasser MW, Ganti AK, Salgia R, Kumar S, Jain M, Ponnusamy MP, and Batra SK
- Abstract
Aberrantly expressed onco-mucin 16 (MUC16) and its post-cleavage generated surface tethered carboxy-terminal (MUC16-Cter) domain are strongly associated with poor prognosis and lethality of pancreatic (PC) and non-small cell lung cancer (NSCLC). To date, most anti-MUC16 antibodies are directed towards the extracellular domain of MUC16 (CA125), which is usually cleaved and shed in the circulation hence obscuring antibody accessibility to the cancer cells. Herein, we establish the utility of targeting a post-cleavage generated, surface-tethered oncogenic MUC16 carboxy-terminal (MUC16-Cter) domain by using a novel chimeric antibody in human IgG1 format, ch5E6, whose epitope expression directly correlates with disease severity in both cancers. ch5E6 binds and interferes with MUC16-associated oncogenesis, suppresses the downstream signaling pFAK(Y397)/p-p70S6K(T389)/N-cadherin axis and exert antiproliferative effects in cancer cells, 3D organoids, and tumor xenografts of both PC and NSCLC. The robust clinical correlations observed between MUC16 and N-cadherin in patient tumors and metastatic samples imply ch5E6 potential in targeting a complex and significantly occurring phenomenon of epithelial to mesenchymal transition (EMT) associated with disease aggressiveness. Our study supports evaluating ch5E6 with standard-of-care drugs, to potentially augment treatment outcomes in malignancies inflicted with MUC16-associated poor prognosis., (© 2023. Nature Publishing Group UK.)
- Published
- 2023
- Full Text
- View/download PDF
11. Highly Selective Targeting of Pancreatic Cancer in the Liver with a Near-Infrared Anti-MUC5AC Probe in a PDOX Mouse Model: A Proof-of-Concept Study.
- Author
-
Turner MA, Cox KE, Neel N, Amirfakhri S, Nishino H, Clary BM, Hosseini M, Natarajan G, Mallya K, Mohs AM, Hoffman RM, Batra SK, and Bouvet M
- Abstract
Accurately identifying metastatic disease is critical to directing the appropriate treatment in pancreatic cancer. Mucin 5AC is overexpressed in pancreatic cancer but absent in normal pancreas tissue. The present proof-of-concept study demonstrates the efficacy of an anti-mucin 5AC antibody conjugated to an IR800 dye (MUC5AC-IR800) to preferentially label a liver metastasis of pancreatic cancer ( Panc Met ) in a unique patient-derived orthotopic xenograft (PDOX) model. In orthotopic models, the mean tumor to background ratio was 1.787 (SD ± 0.336), and immunohistochemistry confirmed the expression of MUC5AC within tumor cells. MUC5AC-IR800 provides distinct visualization of pancreatic cancer liver metastasis in a PDOX mouse model, demonstrating its potential utility in staging laparoscopy and fluorescence-guided surgery.
- Published
- 2023
- Full Text
- View/download PDF
12. Specific Targeting and Labeling of Colonic Polyps in CPC-APC Mice with Mucin 5AC Fluorescent Antibodies: A Model for Detection of Early Colon Cancer.
- Author
-
Turner MA, Cox KE, Liu S, Neel N, Amirfakhri S, Nishino H, Hosseini M, Alcantara JA, Abd El-Hafeez AA, Lwin TM, Mallya K, Pisegna JR, Singh SK, Ghosh P, Hoffman RM, Batra SK, and Bouvet M
- Abstract
Poor visualization of polyps can limit colorectal cancer screening. Fluorescent antibodies to mucin5AC (MUC5AC), a glycoprotein upregulated in adenomas and colorectal cancer, could improve screening colonoscopy polyp detection rate. Adenomatous polyposis coli flox mice with a Cdx2-Cre transgene ( CPC-APC ) develop colonic polyps that contain both dysplastic and malignant tissue. Mice received MUC5AC-IR800 or IRdye800 as a control IV and were sacrificed after 48 h for near-infrared imaging of their colons. A polyp-to-background ratio (PBR) was calculated for each polyp by dividing the mean fluorescence intensity of the polyp by the mean fluorescence intensity of the background tissue. The mean 25 μg PBR was 1.70 (±0.56); the mean 50 μg PBR was 2.64 (±0.97); the mean 100 μg PBR was 3.32 (±1.33); and the mean 150 μg PBR was 3.38 (±0.87). The mean PBR of the dye-only control was 2.22 (±1.02), significantly less than the 150 μg arm ( p -value 0.008). The present study demonstrates the ability of fluorescent anti-MUC5AC antibodies to specifically target and label colonic polyps containing high-grade dysplasia and intramucosal adenocarcinoma in CPC-APC mice. This technology can potentially improve the detection rate and decrease the miss rate of advanced colonic neoplasia and early cancer at colonoscopy.
- Published
- 2023
- Full Text
- View/download PDF
13. Muc16 depletion diminishes KRAS-induced tumorigenesis and metastasis by altering tumor microenvironment factors in pancreatic ductal adenocarcinoma.
- Author
-
Lakshmanan I, Marimuthu S, Chaudhary S, Seshacharyulu P, Rachagani S, Muniyan S, Chirravuri-Venkata R, Atri P, Rauth S, Nimmakayala RK, Siddiqui JA, Gautam SK, Shah A, Natarajan G, Parte S, Bhyravbhatla N, Mallya K, Haridas D, Talmon GA, Smith LM, Kumar S, Ganti AK, Jain M, Ponnusamy MP, and Batra SK
- Subjects
- Animals, Mice, Carcinogenesis, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Tumor Microenvironment genetics, Carcinoma, Pancreatic Ductal pathology, Mucins metabolism, Pancreatic Neoplasms pathology
- Abstract
MUC16, membrane-bound mucin, plays an oncogenic role in pancreatic ductal adenocarcinoma (PDAC). However, the pathological role of MUC16 in the PDAC progression, tumor microenvironment, and metastasis in cooperation with Kras
G12D and Trp53R172H mutations remains unknown. Deletion of Muc16 with activating mutations KrasG12D/+ and Trp53R172H/+ in mice significantly decreased progression and prolonged overall survival in KrasG12D/+ ; Trp53R172H/+ ; Pdx-1-Cre; Muc16-/- (KPCM) and KrasG12D/+ ; Pdx-1-Cre; Muc16-/- (KCM), as compared to KrasG12D/+ ; Trp53R172H/+ ; Pdx-1-Cre (KPC) and KrasG12D/+ ; Pdx-1-Cre (KC) mice, respectively. Muc16 knockout pancreatic tumor (KPCM) displays decreased tumor microenvironment factors and significantly reduced incidence of liver and lung metastasis compared to KPC. Furthermore, in silico data analysis showed a positive correlation of MUC16 with activated stroma and metastasis-associated genes. KPCM mouse syngeneic cells had significantly lower metastatic and endothelial cell binding abilities than KPC cells. Similarly, KPCM organoids significantly decreased the growth rate compared to KPC organoids. Interestingly, RNA-seq data revealed that the cytoskeletal proteins Actg2, Myh11, and Pdlim3 were downregulated in KPCM tumors. Further knockdown of these genes showed reduced metastatic potential. Overall, our results demonstrate that Muc16 alters the tumor microenvironment factors during pancreatic cancer progression and metastasis by changing the expression of Actg2, Myh11, and Pdlim3 genes., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
14. Anti-mucin 4 fluorescent antibody brightly targets colon cancer in patient-derived orthotopic xenograft mouse models: A proof-of-concept study for future clinical applications.
- Author
-
Turner MA, Hollandsworth HM, Amirfakhri S, Lwin TM, Nishino H, Neel NC, Natarajan G, Kaur S, Mallya K, Hoffman RM, Batra SK, and Bouvet M
- Subjects
- Animals, Disease Models, Animal, Heterografts, Humans, Mice, Mice, Nude, Colonic Neoplasms surgery, Liver Neoplasms surgery
- Abstract
Background: There is a high rate of positive surgical margins with resection of liver metastases in colorectal cancer (CRC). The present study reports using a fluorescent anti-mucin 4 (MUC4) antibodies to label primary CRC and liver metastases to better visualize tumor margins in mouse models., Methods: Western blotting for MUC4 protein expression of normal colon and CRC tumor lysates was performed. Orthotopic primary and liver metastatic CRC mouse models received anti-MUC4 antibody conjugated to IR800 (MUC4-IR800). Mice were sacrificed and imaged after 48 hours., Results: Western blotting demonstrated increased MUC4 expression in a human CRC cell line and patient-derived primary and liver-metastatic CRCs. The LS174T orthotopic primary CRC model tumor to background ratio (TBR) was 2.04 (±0.35). The patient-derived orthotopic xenograft (PDOX) primary CRC model TBR was 2.17 (±0.35). The PDOX liver metastasis model TBR was 1.56 (±0.53)., Conclusion: MUC4-IR800 provided bright labeling of primary and liver tumors in CRC orthotopic mouse models, demonstrating their future clinical potential for margin visualization in fluorescence guided surgery., Competing Interests: Declaration of competing interest RMH is a non-salaried affiliate of AntiCancer Inc. which uses PDOX models for contract research. All other authors have nothing to disclose., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
15. MUC16 Promotes Liver Metastasis of Pancreatic Ductal Adenocarcinoma by Upregulating NRP2-Associated Cell Adhesion.
- Author
-
Marimuthu S, Lakshmanan I, Muniyan S, Gautam SK, Nimmakayala RK, Rauth S, Atri P, Shah A, Bhyravbhatla N, Mallya K, Grandgenett PM, Hollingsworth MA, Datta K, Jain M, Ponnusamy MP, and Batra SK
- Subjects
- Animals, CA-125 Antigen metabolism, Cell Adhesion, Cell Line, Tumor, Cell Movement, Cell Proliferation, Humans, Membrane Proteins metabolism, Mice, Neoplasm Metastasis, Pancreatic Neoplasms, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology, Liver Neoplasms genetics, Liver Neoplasms secondary, Neuropilin-2 genetics, Pancreatic Neoplasms pathology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal types of cancer, as it commonly metastasizes to the liver resulting in an overall poor prognosis. However, the molecular mechanism involved in liver metastasis remains poorly understood. Here, we aimed to identify the MUC16-mediated molecular mechanism of PDAC-liver metastasis. Previous studies demonstrated that MUC16 and its C-terminal (Cter) domain are involved in the aggressiveness of PDAC. In this study, we observed MUC16 and its Cter expression significantly high in human PDAC tissues, PDAC organoids, and metastatic liver tissues, while no expression was observed in normal pancreatic tissues using IHC and immunofluorescence (IFC) analyses. MUC16 knockdown in SW1990 and CD18/HPAF PDAC cells significantly decreased the colony formation, migration, and endothelial/p-selectin binding. In contrast, MUC16-Cter ectopic overexpression showed significantly increased colony formation and motility in MiaPaCa2 pancreatic cancer cells. Interestingly, MUC16 promoted cell survival and colonization in the liver, mimicking an ex vivo environment. Furthermore, MUC16 enhanced liver metastasis in the in vivo mouse model. Our integrated analyses of RNA-sequencing suggested that MUC16 alters Neuropilin-2 (NRP2) and cell adhesion molecules in pancreatic cancer cells. Furthermore, we identified that MUC16 regulated NRP2 via JAK2/STAT1 signaling in PDAC. NRP2 knockdown in MUC16-overexpressed PDAC cells showed significantly decreased cell adhesion and migration. Overall, the findings indicate that MUC16 regulates NRP2 and induces metastasis in PDAC., Implications: This study shows that MUC16 plays a critical role in PDAC liver metastasis by mediating NRP2 regulation by JAK2/STAT1 axis, thereby paving the way for future therapy efforts for metastatic PDAC., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
16. DNA-gold nanoprobe-based integrated biosensing technology for non-invasive liquid biopsy of serum miRNA: A new frontier in prostate cancer diagnosis.
- Author
-
Kshirsagar P, Seshacharyulu P, Muniyan S, Rachagani S, Smith LM, Thompson C, Shah A, Mallya K, Kumar S, Jain M, and Batra SK
- Subjects
- Animals, Biomarkers, Tumor genetics, Biopsy, DNA, Gold, Humans, Liquid Biopsy, Male, Mice, Prostate-Specific Antigen genetics, Technology, Metal Nanoparticles, MicroRNAs genetics, Prostatic Neoplasms diagnosis, Prostatic Neoplasms genetics
- Abstract
The low specificity of prostate-specific antigen contributes to overdiagnosis and ov ertreatment of prostate cancer (PCa) patients. Hence, there is an urgent need for inclusive diagnostic platforms that could improve the diagnostic accuracy of PCa. Dysregulated miRNAs are closely associated with the progression and recurrence and have emerged as promising diagnostic and prognostic biomarkers for PCa. Nevertheless, simple, rapid, and ultrasensitive quantification of serum miRNAs is highly challenging. This study designed, synthesized, and demonstrated the practicability of DNA-linked gold nanoprobes (DNA-AuNPs) for the single-step quantification of miR-21/miR-141/miR-375. In preclinical study, the assay differented PCa Pten conditional knockout (Pten
cKO ) mice compared to their age-matched Pten wild-type (PtenWT ) control mice. In human sera, receiver operating characteristic (ROC) curve-based correlation analyses revealed clear discrimination between PCa patients from normal healthy controls using training and validation sets. Overall, we established integrated nano-biosensing technology for the PCR-free, non-invasive liquid biopsies of multiple miRNAs for PCa diagnosis., Competing Interests: Declaration of competing interest SKB is a founding member of Sanguine Diagnostics and Therapeutics, Inc. All other authors declare no competing financial interest., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
17. Ubiquitous Aberration in Cholesterol Metabolism across Pancreatic Ductal Adenocarcinoma.
- Author
-
Gunda V, Genaro-Mattos TC, Kaushal JB, Chirravuri-Venkata R, Natarajan G, Mallya K, Grandgenett PM, Mirnics K, Batra SK, Korade Z, and Rachagani S
- Abstract
Pancreatic cancer (PC) is characterized by metabolic deregulations that often manifest as deviations in metabolite levels and aberrations in their corresponding metabolic genes across the clinical specimens and preclinical PC models. Cholesterol is one of the critical metabolites supporting PC, synthesized or acquired by PC cells. Nevertheless, the significance of the de novo cholesterol synthesis pathway has been controversial in PC, indicating the need to reassess this pathway in PC. We utilized preclinical models and clinical specimens of PC patients and cell lines and utilized mass spectrometry-based sterol analysis. Further, we also performed in silico analysis to corroborate the significance of de novo cholesterol synthesis pathway in PC. Our results demonstrated alteration in free sterol levels, including free cholesterol, across in vitro, in vivo, and clinical specimens of PC. Especially, our sterol analyses established consistent alterations in free cholesterol across the different PC models. Overall, this study demonstrates the significance and consistency in deviation of cholesterol synthesis pathway in PC while showing the aberrations in sterol metabolite intermediates and the related genes using preclinical models, in silico platforms, and the clinical specimens.
- Published
- 2022
- Full Text
- View/download PDF
18. Fluorescent Anti-MUC5AC Brightly Targets Pancreatic Cancer in a Patient-derived Orthotopic Xenograft.
- Author
-
Turner MA, Hollandsworth HM, Nishino H, Amirfakhri S, Lwin TM, Lowy AM, Kaur S, Natarajan G, Mallya K, Hoffman RM, Batra SK, and Bouvet M
- Subjects
- Animals, Fluorescent Dyes, Heterografts, Humans, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Pancreatic Neoplasms genetics
- Abstract
Background: Overexpression of mucin-5AC (MUC5AC) makes it a targetable biomarker in pancreatic cancer. The present study evaluated tumor targeting with a MUC5AC antibody conjugated to a near-infrared dye in a patient-derived orthotopic xenograft (PDOX) mouse model., Materials and Methods: MUC5AC monoclonal antibody was conjugated to the near-infrared dye IRDye800CW to synthesize MUC5AC-IR800. PDOX models were established by implanting a high-MUC5AC-expressing patient-derived pancreatic tumor on the pancreas of nude mice. After 4 weeks of PDOX tumor growth, mice were imaged after receiving MUC5AC-IR800 (75 μg) intravenously., Results: In the PDOX models, MUC5AC-IR800 selectively and brightly targeted the pancreatic tumor (tumor to background ratio: 2.46±0.465)., Conclusion: MUC5AC-IR800 provides distinct visualization of pancreatic tumors. MUC5AC-IR800 may be used clinically in the future to improve pancreatic cancer resection. This novel fluorescent probe is also promising for targeting of pre-malignant pancreatic lesions with subsequent resection under fluorescence guidance., (Copyright © 2022 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Mucin 5AC Serves as the Nexus for β-Catenin/c-Myc Interplay to Promote Glutamine Dependency During Pancreatic Cancer Chemoresistance.
- Author
-
Ganguly K, Bhatia R, Rauth S, Kisling A, Atri P, Thompson C, Vengoji R, Ram Krishn S, Shinde D, Thomas V, Kaur S, Mallya K, Cox JL, Kumar S, and Batra SK
- Subjects
- Animals, Cell Line, Tumor, Databases, Genetic, Deoxycytidine pharmacology, Enzyme Inhibitors pharmacology, Female, Gene Expression Regulation, Neoplastic, Glutaminase antagonists & inhibitors, Glutaminase metabolism, Humans, Male, Mice, Knockout, Mice, Nude, Mucin 5AC genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins c-myc genetics, Signal Transduction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, beta Catenin antagonists & inhibitors, beta Catenin genetics, Gemcitabine, Mice, Antimetabolites, Antineoplastic pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Deoxycytidine analogs & derivatives, Drug Resistance, Neoplasm genetics, Energy Metabolism drug effects, Glutamine metabolism, Mucin 5AC metabolism, Pancreatic Neoplasms drug therapy, Proto-Oncogene Proteins c-myc metabolism, beta Catenin metabolism
- Abstract
Background & Aims: A major clinical challenge for patients with pancreatic cancer (PC) is metabolic adaptation. Neoplastic cells harboring molecular perturbations suffice for their increased anabolic demand and nucleotide biosynthesis to acquire chemoresistance. The mucin 5AC expressed de novo in malignant pancreas promotes cancer cell stemness and is significantly associated with poor patient survival. Identification of MUC5AC-associated drivers of chemoresistance through metabolic alterations may facilitate the sculpting of a new combinatorial regimen., Methods: The contributions of MUC5AC to glutaminolysis and gemcitabine resistance were examined by The Cancer Genome Atlas data analysis, RNA sequencing, and immunohistochemistry analysis on pancreatic tissues of Kras
G12D ;Pdx1-Cre (KC) and KrasG12D ;Pdx1-Cre;Muc5ac-/- mice. These were followed by metabolite flux assays as well as biochemical and xenograft studies on MUC5AC-depleted human and murine PC cells. Murine and human pancreatic 3-dimensional tumoroids were used to evaluate the efficacy of gemcitabine in combination with β-catenin and glutaminolysis inhibitors., Results: Transcriptional analysis showed that high MUC5AC-expressing human and autochthonous murine PC tumors exhibit higher resistance to gemcitabine because of enhanced glutamine use and nucleotide biosynthesis. Gemcitabine treatment led to MUC5AC overexpression, resulting in disruption of E-cadherin/β-catenin junctions and the nuclear translocation of β-catenin, which increased c-Myc expression, with a concomitant rise in glutamine uptake and glutamate release. MUC5AC depletion and glutamine deprivation sensitized human PC cells to gemcitabine, which was obviated by glutamine replenishment in MUC5AC-expressing cells. Coadministration of β-catenin and glutaminolysis inhibitors with gemcitabine abrogated the MUC5AC-mediated resistance in murine and human tumoroids., Conclusions: The MUC5AC/β-catenin/c-Myc axis increases the uptake and use of glutamine in PC cells, and cotargeting this axis along with gemcitabine may improve therapeutic efficacy in PC., (Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
20. Reduction in O-glycome induces differentially glycosylated CD44 to promote stemness and metastasis in pancreatic cancer.
- Author
-
Leon F, Seshacharyulu P, Nimmakayala RK, Chugh S, Karmakar S, Nallasamy P, Vengoji R, Rachagani S, Cox JL, Mallya K, Batra SK, and Ponnusamy MP
- Subjects
- Cell Differentiation, Cell Line, Tumor, Glycosylation, Humans, Neoplasm Metastasis, Neoplastic Stem Cells pathology, Pancreatic Neoplasms pathology, Hyaluronan Receptors metabolism, Neoplastic Stem Cells physiology, Pancreatic Neoplasms genetics
- Abstract
Aberrant protein glycosylation has been shown to have a significant contribution in aggressive cancer, including pancreatic cancer (PC). Emerging evidence has implicated the involvement of cancer stem cells (CSCs) in PC aggressiveness; however, the contribution of glycosylation on self-renewal properties and maintenance of CSC is understudied. Here, using several in vitro and in vivo models lacking C1GALT1 expression, we identified the role of aberrant O-glycosylation in stemness properties and aggressive PC metastasis. A loss in C1GALT1 was found to result in the truncation of O-glycosylation on several glycoproteins with an enrichment of Tn carbohydrate antigen. Mapping of Tn-bearing glycoproteins in C1GALT1 KO cells identified significant Tn enrichment on CSC glycoprotein CD44. Notably, a loss of C1GALT1 in PC cells was found to enhance CSC features (side population-SP, ALDH1+, and tumorspheres) and self-renewal markers NANOG, SOX9, and KLF4. Furthermore, a loss of CD44 in existing C1GALT1 KO cells decreased NANOG expression and CSC features. We determined that O-glycosylation of CD44 activates ERK/NF-kB signaling, which results in increased NANOG expression in PC cells that facilitated the alteration of CSC features, suggesting that NANOG is essential for PC stemness. Finally, we identified that loss of C1GALT1 expression was found to augment tumorigenic and metastatic potential, while an additional loss of CD44 in these cells reversed the effects. Overall, our results identified that truncation of O-glycans on CD44 increases NANOG activation that mediates increased CSC activation., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
21. Characterization of recombinant β subunit of human MUC4 mucin (rMUC4β).
- Author
-
Kshirsagar PG, Gulati M, Junker WM, Aithal A, Spagnol G, Das S, Mallya K, Gautam SK, Kumar S, Sorgen P, Pandey KK, Batra SK, and Jain M
- Subjects
- Cloning, Molecular, Gene Expression, Gene Order, Humans, Mass Spectrometry, Models, Molecular, Mucin-4 chemistry, Mucin-4 isolation & purification, Plasmids genetics, Protein Binding, Structure-Activity Relationship, Mucin-4 genetics, Mucin-4 metabolism, Protein Subunits, Recombinant Proteins
- Abstract
MUC4 is a transmembrane mucin expressed on various epithelial surfaces, including respiratory and gastrointestinal tracts, and helps in their lubrication and protection. MUC4 is also aberrantly overexpressed in various epithelial malignancies and functionally contributes to cancer development and progression. MUC4 is putatively cleaved at the GDPH site into a mucin-like α-subunit and a membrane-tethered growth factor-like β-subunit. Due to the presence of several functional domains, the characterization of MUC4β is critical for understanding MUC4 biology. We developed a method to produce and purify multi-milligram amounts of recombinant MUC4β (rMUC4β). Purified rMUC4β was characterized by Far-UV CD and I-TASSER-based protein structure prediction analyses, and its ability to interact with cellular proteins was determined by the affinity pull-down assay. Two of the three EGF-like domains exhibited typical β-fold, while the third EGF-like domain and vWD domain were predominantly random coils. We observed that rMUC4β physically interacts with Ezrin and EGFR family members. Overall, this study describes an efficient and simple strategy for the purification of biologically-active rMUC4β that can serve as a valuable reagent for a variety of biochemical and functional studies to elucidate MUC4 function and generating domain-specific antibodies and vaccines for cancer immunotherapy., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. Pancreatic Tumor Microenvironment Factor Promotes Cancer Stemness via SPP1-CD44 Axis.
- Author
-
Nallasamy P, Nimmakayala RK, Karmakar S, Leon F, Seshacharyulu P, Lakshmanan I, Rachagani S, Mallya K, Zhang C, Ly QP, Myers MS, Josh L, Grabow CE, Gautam SK, Kumar S, Lele SM, Jain M, Batra SK, and Ponnusamy MP
- Subjects
- Animals, Cancer-Associated Fibroblasts pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Movement, Cell Proliferation, Coculture Techniques, Epithelial-Mesenchymal Transition, Gene Expression Regulation, Neoplastic, Humans, Hyaluronan Receptors genetics, Male, Mice, Nude, Mice, Transgenic, Neoplasm Invasiveness, Neoplastic Stem Cells pathology, Osteopontin genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Paracrine Communication, Phenotype, Signal Transduction, Mice, Cancer-Associated Fibroblasts metabolism, Carcinoma, Pancreatic Ductal metabolism, Hyaluronan Receptors metabolism, Neoplastic Stem Cells metabolism, Osteopontin metabolism, Pancreatic Neoplasms metabolism, Tumor Microenvironment
- Abstract
Background & Aims: Tumor-microenvironment factors and cancer stem cells (CSCs) play a critical role in the aggressiveness of pancreatic cancer (PC). However, the degree to which tumor-microenvironment factors promote stemness remains unexplored. Here, we examined whether cancer-associated fibroblasts (CAFs) promote CSC features in PC., Methods: PC cells were treated long-term (30, 60, and 90 days) with conditioned media (CM)-derived from normal human fibroblasts (NFs) and CAFs. The stemness features of tumorsphere formation and stemness populations, along with CSCs markers, were analyzed using 2-dimensional and 3-dimensional sodium alginate bead-based co-culture models. Immunohistochemistry and immunofluorescence staining were performed for CSCs and fibroblast markers in autochthonous Kras
G12D/+ ; Trp53R172H/+ ; Pdx1-Cre mice and human pancreatic tumors. Polymerase chain reaction array and gene knockdown were performed to identify the mechanism of stemness enrichment., Results: Long-term treatment of PC cells with CAF-CM enriched stemness, as indicated by significantly higher CD44+ , ALDH+ , and AF+ populations in PC cells. Increased tumorsphere formation and elevated CSC, self-renewal, and drug-resistance markers in CAF-CM-treated PC cells were observed. In addition, CAFs co-cultured with PC cells in the 3-dimensional model showed a substantial increase in stemness features. CD44 and α-smooth muscle actin were positively correlated and their expressions progressively increased from the early to late stages of KrasG12D/+ ; Trp53R172H/+ ; Pdx1-Cre mouse and human pancreatic tumors. Osteopontin/secreted phosphoprotein 1 was identified as the top differentially overexpressed gene in CAF-CM-treated PC cells and knockdown of osteopontin/secreted phosphoprotein 1 significantly reduced stemness characteristics in CAF-CM-treated PC cells., Conclusions: Our data uncovered novel insight into the interplay between CAF and enrichment of stemness population through the osteopontin/secreted phosphoprotein 1-CD44 axis in PC., (Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
23. Differential gene expression-based connectivity mapping identified novel drug candidate and improved Temozolomide efficacy for Glioblastoma.
- Author
-
Vengoji R, Atri P, Macha MA, Seshacharyulu P, Perumal N, Mallya K, Liu Y, Smith LM, Rachagani S, Mahapatra S, Ponnusamy MP, Jain M, Batra SK, and Shonka N
- Subjects
- Animals, Antineoplastic Agents, Alkylating therapeutic use, Cell Line, Tumor, Cell Survival, Computational Biology methods, DNA Breaks, Disease Management, Disease Models, Animal, Dose-Response Relationship, Drug, Drug Discovery methods, Drug Resistance, Neoplasm genetics, Gene Expression Profiling, Glioblastoma diagnosis, Glioblastoma drug therapy, Humans, Mice, Mice, Transgenic, Temozolomide therapeutic use, Transcriptome, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Temozolomide pharmacology
- Abstract
Background: Glioblastoma (GBM) has a devastating median survival of only one year. Treatment includes resection, radiation therapy, and temozolomide (TMZ); however, the latter increased median survival by only 2.5 months in the pivotal study. A desperate need remains to find an effective treatment., Methods: We used the Connectivity Map (CMap) bioinformatic tool to identify candidates for repurposing based on GBM's specific genetic profile. CMap identified histone deacetylase (HDAC) inhibitors as top candidates. In addition, Gene Expression Profiling Interactive Analysis (GEPIA) identified HDAC1 and HDAC2 as the most upregulated and HDAC11 as the most downregulated HDACs. We selected PCI-24781/abexinostat due to its specificity against HDAC1 and HDAC2, but not HDAC11, and blood-brain barrier permeability., Results: We tested PCI-24781 using in vitro human and mouse GBM syngeneic cell lines, an in vivo murine orthograft, and a genetically engineered mouse model for GBM (PEPG - PTEN
flox/+ ; EGFRvIII+; p16Flox/- & GFAP Cre +). PCI-24781 significantly inhibited tumor growth and downregulated DNA repair machinery (BRCA1, CHK1, RAD51, and O6 -methylguanine-DNA- methyltransferase (MGMT)), increasing DNA double-strand breaks and causing apoptosis in the GBM cell lines, including an MGMT expressing cell line in vitro. Further, PCI-24781 decreased tumor burden in a PEPG GBM mouse model. Notably, TMZ + PCI increased survival in orthotopic murine models compared to TMZ + vorinostat, a pan-HDAC inhibitor that proved unsuccessful in clinical trials., Conclusion: PCI-24781 is a novel GBM-signature specific HDAC inhibitor that works synergistically with TMZ to enhance TMZ efficacy and improve GBM survival. These promising MGMT-agnostic results warrant clinical evaluation., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
24. PGC1α-Mediated Metabolic Reprogramming Drives the Stemness of Pancreatic Precursor Lesions.
- Author
-
Nimmakayala RK, Rauth S, Chirravuri Venkata R, Marimuthu S, Nallasamy P, Vengoji R, Lele SM, Rachagani S, Mallya K, Malafa MP, Ponnusamy MP, and Batra SK
- Subjects
- Animals, Humans, Mice, Pancreas pathology, Pancreatic Ducts pathology, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Purpose: Metabolic reprogramming and cancer stem cells drive the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). However, the metabolic and stemness programs of pancreatic precursor lesions (PPL), considered early PDAC development events, have not been thoroughly explored., Experimental Design: Meta-analyses using gene expression profile data from NCBI Gene Expression Omnibus and IHC on tissue microarrays (TMA) were performed. The following animal and cellular models were used: cerulean-induced KrasG12D; Pdx1 Cre (KC) acinar-to-ductal metaplasia (ADM) mice, KrasG12D; Smad4Loss; Pdx-1 Cre (KCSmad4-) intraductal papillary mucinous neoplasm (IPMN) mice, LGKC1 cell line derived from the doxycycline-inducible Gnas IPMN model, and human IPMN organoids. Flow cytometry, Seahorse extracellular flux analyzer, qRT-PCR, and sphere assay were used to analyze metabolic and stemness features. SR18292 was used to inhibit PGC1α, and short hairpin RNA was used to knockdown (KD) PGC1α., Results: The meta-analysis revealed a significant upregulation of specific stemness genes in ADM-mediated pancreatic intraepithelial neoplasms (PanIN) and IPMN. Meta- and TMA analyses followed by in vitro and in vivo validation revealed that ADM/PanIN exhibit increased PGC1α and oxidative phosphorylation (OXPhos) but reduced CPT1A. IPMN showed elevated PGC1α, fatty acid β-oxidation (FAO) gene expression, and FAO-OXPhos. PGC1α was co-overexpressed with its coactivator NRF1 in ADM/PanINs and with PPARγ in IPMN. PGC1α KD or SR18292 inhibited the specific metabolic and stemness features of PPLs and repressed IPMN organoid growth., Conclusions: ADM/PanINs and IPMNs show specific stemness signatures with unique metabolisms. Inhibition of PGC1α using SR18292 diminishes the specific stemness by targeting FAO-independent and FAO-dependent OXPhos of ADM/PanINs and IPMNs, respectively., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
25. Modeling pancreatic cancer in mice for experimental therapeutics.
- Author
-
Mallya K, Gautam SK, Aithal A, Batra SK, and Jain M
- Subjects
- Animals, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Drug Resistance, Neoplasm, Genetic Predisposition to Disease, Mice, Transgenic, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Phenotype, Species Specificity, Tumor Microenvironment, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Carcinoma, Pancreatic Ductal therapy, Immunotherapy, Neoplasms, Experimental therapy, Pancreatic Neoplasms therapy
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. Dual blockade of EGFR and CDK4/6 delays head and neck squamous cell carcinoma progression by inducing metabolic rewiring.
- Author
-
Chaudhary S, Pothuraju R, Rachagani S, Siddiqui JA, Atri P, Mallya K, Nasser MW, Sayed Z, Lyden ER, Smith L, Gupta SD, Ralhan R, Lakshmanan I, Jones DT, Ganti AK, Macha MA, and Batra SK
- Subjects
- Animals, Disease Models, Animal, Disease Progression, ErbB Receptors antagonists & inhibitors, Humans, Mice, Squamous Cell Carcinoma of Head and Neck pathology, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Despite preclinical success, monotherapies targeting EGFR or cyclin D1-CDK4/6 in Head and Neck squamous cell carcinoma (HNSCC) have shown a limited clinical outcome. Here, we aimed to determine the combined effect of palbociclib (CDK4/6) and afatinib (panEGFR) inhibitors as an effective strategy to target HNSCC. Using TCGA-HNSCC co-expression analysis, we found that patients with high EGFR and cyclin D1 expression showed enrichment of gene clusters associated with cell-growth, glycolysis, and epithelial to mesenchymal transition processes. Phosphorylated S6 (p-S6), a downstream effector of EGFR and cyclin D1-CDK4/6 signalling, showed a progressive increase from normal oral tissues to leukoplakia and frank malignancy, and associated with poor outcome of the patients. This increased p-S6 expression was drastically reduced after combination treatment with afatinib and palbociclib in the cell lines and mouse models, suggesting its utiliy as a prognostic marker in HNSCC. Combination treatment also reduced the cell growth and induced cell senescence via increasing reactive oxygen species with concurrent ablation of glycolytic and tricarboxylic acid cycle intermediates. Finally, our findings in sub-cutaneous and genetically engineered mouse model (K14-CreER
tam ;LSL-KrasG12D /+ ;Trp53R172H /+ ) studies showed a significant reduction in the tumor growth and delayed tumor progression after combination treatment. This study collectively demonstrates that dual targeting may be a critical therapeutic strategy in blocking tumor progression via inducing metabolic alteration and warrants clinical evaluation., (Copyright © 2021 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
27. ST6GalNAc-I promotes lung cancer metastasis by altering MUC5AC sialylation.
- Author
-
Lakshmanan I, Chaudhary S, Vengoji R, Seshacharyulu P, Rachagani S, Carmicheal J, Jahan R, Atri P, Chirravuri-Venkata R, Gupta R, Marimuthu S, Perumal N, Rauth S, Kaur S, Mallya K, Smith LM, Lele SM, Ponnusamy MP, Nasser MW, Salgia R, Batra SK, and Ganti AK
- Subjects
- Animals, Glycosylation, Humans, Mice, Mucin 5AC metabolism, N-Acetylneuraminic Acid, Sialyltransferases genetics, Sialyltransferases metabolism, Liver Neoplasms, Lung Neoplasms genetics
- Abstract
Lung cancer (LC) is the leading cause of cancer-related mortality. However, the molecular mechanisms associated with the development of metastasis are poorly understood. Understanding the biology of LC metastasis is critical to unveil the molecular mechanisms for designing targeted therapies. We developed two genetically engineered LC mouse models Kras
G12D/+ ; Trp53R172H/+ ; Ad-Cre (KPA) and KrasG12D/+ ; Ad-Cre (KA). Survival analysis showed significantly (P = 0.0049) shorter survival in KPA tumor-bearing mice as compared to KA, suggesting the aggressiveness of the model. Our transcriptomic data showed high expression of N-acetylgalactosaminide alpha-2, 6-sialyltransferase 1 (St6galnac-I) in KPA compared to KA tumors. ST6GalNAc-I is an O-glycosyltransferase, which catalyzes the addition of sialic acid to the initiating GalNAc residues forming sialyl Tn (STn) on glycoproteins, such as mucins. Ectopic expression of species-specific p53 mutants in the syngeneic mouse and human LC cells led to increased cell migration and high expression of ST6GalNAc-I, STn, and MUC5AC. Immunoprecipitation of MUC5AC in the ectopically expressing p53R175H cells exhibited higher affinity toward STn. In addition, ST6GalNAc-I knockout (KO) cells also showed decreased migration, possibly due to reduced glycosylation of MUC5AC as observed by low STn on the glycoprotein. Interestingly, ST6GalNAc-I KO cells injected mice developed less liver metastasis (P = 0.01) compared to controls, while colocalization of MUC5AC and STn was observed in the liver metastatic tissues of control mice. Collectively, our findings support the hypothesis that mutant p53R175H mediates ST6GalNAc-I expression, leading to the sialyation of MUC5AC, and thus contribute to LC liver metastasis., (© 2021 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2021
- Full Text
- View/download PDF
28. Metabolic programming of distinct cancer stem cells promotes metastasis of pancreatic ductal adenocarcinoma.
- Author
-
Nimmakayala RK, Leon F, Rachagani S, Rauth S, Nallasamy P, Marimuthu S, Shailendra GK, Chhonker YS, Chugh S, Chirravuri R, Gupta R, Mallya K, Prajapati DR, Lele SM, C Caffrey T, L Grem J, Grandgenett PM, Hollingsworth MA, Murry DJ, Batra SK, and Ponnusamy MP
- Subjects
- AC133 Antigen metabolism, ATP Binding Cassette Transporter, Subfamily B metabolism, Animals, Carcinoma, Pancreatic Ductal drug therapy, Cell Line, Tumor, Coculture Techniques, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Drug Resistance, Neoplasm, Epithelial-Mesenchymal Transition, Gene Expression Regulation, Neoplastic, Glycolysis, Humans, Liver Neoplasms metabolism, Lung Neoplasms metabolism, Metabolomics methods, Mice, Neoplasm Metastasis, Neoplasm Transplantation, Pancreatic Neoplasms drug therapy, Gemcitabine, Aldehyde Dehydrogenase 1 Family metabolism, Carcinoma, Pancreatic Ductal metabolism, L-Lactate Dehydrogenase metabolism, Liver Neoplasms secondary, Lung Neoplasms secondary, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms metabolism, Retinal Dehydrogenase metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) metastasizes to distant organs, which is the primary cause of mortality; however, specific features mediating organ-specific metastasis remain unexplored. Emerging evidence demonstrates that cancer stem cells (CSCs) and cellular metabolism play a pivotal role in metastasis. Here we investigated the role of distinct subtypes of pancreatic CSCs and their metabolomic signatures in organ-specific metastatic colonization. We found that PDAC consists of ALDH+/CD133+ and drug-resistant (MDR1+) subtypes of CSCs with specific metabolic and stemness signatures. Human PDAC tissues with gemcitabine treatment, autochthonous mouse tumors from Kras
G12D ; Pdx1-Cre (KC) and KrasG12D ; Trp53R172H ; Pdx-1 Cre (KPC) mice, and KPC- Liver/Lung metastatic cells were used to evaluate the CSC, EMT (epithelial-to-mesenchymal transition), and metabolic profiles. A strong association was observed between distinct CSC subtypes and organ-specific colonization. The liver metastasis showed drug-resistant CSC- and EMT-like phenotype with aerobic glycolysis and fatty acid β-oxidation-mediated oxidative (glyco-oxidative) metabolism. On the contrary, lung metastasis displayed ALDH+/CD133+ and MET-like phenotype with oxidative metabolism. These results were obtained by evaluating FACS-based side population (SP), autofluorescence (AF+) and Alde-red assays for CSCs, and Seahorse-based oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and fatty acid β-oxidation (FAO)-mediated OCR assays for metabolic features along with specific gene signatures. Further, we developed in vitro human liver and lung PDAC metastasis models by using a combination of liver or lung decellularized scaffolds, a co-culture, and a sphere culture methods. PDAC cells grown in the liver-mimicking model showed the enrichment of MDR1+ and CPT1A+ populations, whereas the PDAC cells grown in the lung-mimicking environment showed the enrichment of ALDH+/CD133+ populations. In addition, we observed significantly elevated expression of ALDH1 in lung metastasis and MDR1/LDH-A expression in liver metastasis compared to human primary PDAC tumors. Our studies elucidate that distinct CSCs adapt unique metabolic signatures for organotropic metastasis, which will pave the way for the development of targeted therapy for PDAC metastasis.- Published
- 2021
- Full Text
- View/download PDF
29. Selective inhibition of stemness through EGFR/FOXA2/SOX9 axis reduces pancreatic cancer metastasis.
- Author
-
Kaushik G, Seshacharyulu P, Rauth S, Nallasamy P, Rachagani S, Nimmakayala RK, Vengoji R, Mallya K, Chirravuri-Venkata R, Singh AB, Foster JM, Ly QP, Smith LM, Lele SM, Malafa MP, Jain M, Ponnusamy MP, and Batra SK
- Subjects
- Afatinib therapeutic use, Animals, Cell Line, Tumor, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, ErbB Receptors antagonists & inhibitors, ErbB Receptors physiology, Hepatocyte Nuclear Factor 3-beta physiology, Humans, Mice, Neoplasm Metastasis, Neoplastic Stem Cells drug effects, Pancreatic Neoplasms pathology, SOX9 Transcription Factor physiology, Gemcitabine, Hepatocyte Nuclear Factor 3-beta antagonists & inhibitors, Pancreatic Neoplasms drug therapy, SOX9 Transcription Factor antagonists & inhibitors
- Abstract
Pancreatic cancer (PC) is difficult to defeat due to mechanism (s) driving metastasis and drug resistance. Cancer stemness is a major challenging phenomenon associated with PC metastasis and limiting therapy efficacy. In this study, we evaluated the pre-clinical and clinical significance of eradicating pancreatic cancer stem cells (PCSC) and its components using a pan-EGFR inhibitor afatinib in combination with gemcitabine. Afatinib in combination with gemcitabine significantly reduced Kras
G12D/+ ; Pdx-1 Cre (KC) (P < 0.01) and KrasG12D/+ ; p53R172H/+ ; Pdx-1 Cre (KPC) (P < 0.05) derived mouse tumoroids and KPC-derived murine syngeneic cell line growth compared to gemcitabine/afatinib alone treatment. The drug combination also reduced PC xenograft tumor burden (P < 0.05) and the incidence of metastasis by affecting key stemness markers, as confirmed by co-localization studies. Moreover, the drug combination significantly decreases the growth of various PC patient-derived organoids (P < 0.001). We found that SOX9 is significantly overexpressed in high-grade PC tumors (P < 0.05) and in chemotherapy-treated patients compared to chemo-naïve patients (P < 0.05). These results were further validated using publicly available datasets. Moreover, afatinib alone or in combination with gemcitabine decreased stemness and tumorspheres by reducing phosphorylation of EGFR family proteins, ERK, FAK, and CSC markers. Mechanistically, afatinib treatment decreased CSC markers by downregulating SOX9 via FOXA2. Indeed, EGFR and FOXA2 depletion reduced SOX9 expression in PCSCs. Taken together, pan-EGFR inhibition by afatinib impedes PCSCs growth and metastasis via the EGFR/ERK/FOXA2/SOX9 axis. This novel mechanism of pan-EGFR inhibitor and its ability to eradicate CSC may serve as a tailor-made approach to enhance chemotherapeutic benefits in other cancer types.- Published
- 2021
- Full Text
- View/download PDF
30. Sildenafil Potentiates the Therapeutic Efficacy of Docetaxel in Advanced Prostate Cancer by Stimulating NO-cGMP Signaling.
- Author
-
Muniyan S, Rachagani S, Parte S, Halder S, Seshacharyulu P, Kshirsagar P, Siddiqui JA, Vengoji R, Rauth S, Islam R, Mallya K, Datta K, Xi L, Das A, Teply BA, Kukreja RC, and Batra SK
- Subjects
- Androgens genetics, Animals, Apoptosis drug effects, Cell Line, Tumor, Cyclic AMP genetics, Cyclic Nucleotide Phosphodiesterases, Type 5 genetics, Disease Models, Animal, Docetaxel adverse effects, Drug Synergism, Epithelial Cells drug effects, Gene Expression Regulation, Neoplastic drug effects, Heterografts, Humans, Male, Mice, Mice, Knockout, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Signal Transduction drug effects, Sildenafil Citrate adverse effects, Docetaxel administration & dosage, Nitric Oxide genetics, Prostatic Neoplasms, Castration-Resistant drug therapy, Sildenafil Citrate administration & dosage
- Abstract
Purpose: Docetaxel plays an indispensable role in the management of advanced prostate cancer. However, more than half of patients do not respond to docetaxel, and those good responders frequently experience significant cumulative toxicity, which limits its dose duration and intensity. Hence, a second agent that could increase the initial efficacy of docetaxel and maintain tolerability at biologically effective doses may improve outcomes for patients., Experimental Design: We determined phosphodiesterase 5 (PDE5) expression levels in human and genetically engineered mouse (GEM) prostate tissues and tumor-derived cell lines. Furthermore, we investigated the therapeutic benefits and underlying mechanism of PDE5 inhibitor sildenafil in combination with docetaxel using in vitro , Pten conditional knockout (cKO), derived tumoroid and xenograft prostate cancer models., Results: PDE5 expression was higher in both human and mouse prostate tumors and cancer cell lines compared with normal tissues/cells. In GEM prostate-derived cell lines, PDE5 expression increased from normal prostate (wild-type) epithelial cells to androgen-dependent and castrated prostate-derived cell lines. The addition of physiologically achievable concentrations of sildenafil enhanced docetaxel-induced prostate cancer cell growth inhibition and apoptosis in vitro , reduced murine 3D tumoroid growth, and in vivo tumorigenicity as compared with docetaxel alone. Furthermore, sildenafil enhanced docetaxel-induced NO and cGMP levels thereby augmenting antitumor activity., Conclusions: Our results demonstrate that sildenafil's addition could sensitize docetaxel chemotherapy in prostate cancer cells at much lesser concentration than needed for inducing cell death. Thus, the combinatorial treatment of sildenafil and docetaxel may improve anticancer efficacy and reduce chemotherapy-induced side-effects among patients with advanced prostate cancer., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
31. Blocking c-MET/ERBB1 Axis Prevents Brain Metastasis in ERBB2+ Breast Cancer.
- Author
-
Gautam SK, Kanchan RK, Siddiqui JA, Maurya SK, Rauth S, Perumal N, Atri P, Venkata RC, Mallya K, Mirza S, Ponnusamy MP, Band V, Mahapatra S, Jain M, Batra SK, and Nasser MW
- Abstract
Brain metastasis (BrM) remains a significant cause of cancer-related mortality in epidermal growth factor receptor 2-positive (ERBB2
+ ) breast cancer (BC) patients. We proposed here that a combination treatment of irreversible tyrosine kinase inhibitor neratinib (NER) and the c-MET inhibitor cabozantinib (CBZ) could prevent brain metastasis. To address this, we first tested the combination treatment of NER and CBZ in the brain-seeking ERBB2+ cell lines SKBrM3 and JIMT-1-BR3, and in ERBB2+ organoids that expressed the c-MET/ERBB1 axis. Next, we developed and characterized an orthotopic mouse model of spontaneous BrM and evaluated the therapeutic effect of CBZ and NER in vivo. The combination treatment of NER and CBZ significantly inhibited proliferation and migration in ERBB2+ cell lines and reduced the organoid growth in vitro. Mechanistically, the combination treatment of NER and CBZ substantially inhibited ERK activation downstream of the c-MET/ERBB1 axis. Orthotopically implanted SKBrM3+ cells formed primary tumor in the mammary fat pad and spontaneously metastasized to the brain and other distant organs. Combination treatment with NER and CBZ inhibited primary tumor growth and predominantly prevented BrM. In conclusion, the orthotopic model of spontaneous BrM is clinically relevant, and the combination therapy of NER and CBZ might be a useful approach to prevent BrM in BC.- Published
- 2020
- Full Text
- View/download PDF
32. Acinar transformed ductal cells exhibit differential mucin expression in a tamoxifen-induced pancreatic ductal adenocarcinoma mouse model.
- Author
-
Mallya K, Haridas D, Seshacharyulu P, Pothuraju R, Junker WM, Krishn SR, Muniyan S, Vengoji R, Batra SK, and Rachagani S
- Subjects
- Animals, Biomarkers, Carcinoma, Pancreatic Ductal metabolism, Cell Transformation, Neoplastic metabolism, Disease Models, Animal, Disease Progression, Disease Susceptibility, Gene Expression, Immunohistochemistry, Mice, Mucins metabolism, Multigene Family, Pancreatic Neoplasms metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Transcription Factors, Carcinoma, Pancreatic Ductal etiology, Carcinoma, Pancreatic Ductal pathology, Cell Transformation, Neoplastic chemically induced, Cell Transformation, Neoplastic genetics, Mucins genetics, Pancreatic Neoplasms etiology, Pancreatic Neoplasms pathology, Tamoxifen adverse effects
- Abstract
Pancreatic cancer (PC) is acquired postnatally; to mimic this scenario, we developed an inducible Kras
G12D ; Ptf1a-CreER™ (iKC) mouse model, in which Kras is activated postnatally at week 16 upon tamoxifen (TAM) administration. Upon TAM treatment, iKC mice develop pancreatic intraepithelial neoplasia (PanIN) lesions and PC with metastasis at the fourth and fortieth weeks, respectively, and exhibited acinar-to-ductal metaplasia (ADM) and transdifferentiation. Kras activation upregulated the transcription factors Ncoa3, p-cJun and FoxM1, which in turn upregulated expression of transmembrane mucins (Muc1, Muc4 and Muc16) and secretory mucin (Muc5Ac). Interestingly, knockdown of KrasG12D in multiple PC cell lines resulted in downregulation of MUC1, MUC4, MUC5AC and MUC16. In addition, iKC mice exhibited ADM and transdifferentiation. Our results show that the iKC mouse more closely mimics human PC development and can be used to investigate pancreatic ductal adenocarcinoma (PDAC) biomarkers, early onset of PDAC, and ADM. The iKC model can also be used for preclinical strategies such as targeting mucin axis alone or in combination with neo-adjuvant, immunotherapeutic approaches and to monitor chemotherapy response., Competing Interests: Competing interestsS.K.B. is one of the co-founders for the Sanguine Diagnostics and Therapeutics, Inc. Other co-authors have no competing interests., (© 2020. Published by The Company of Biologists Ltd.)- Published
- 2020
- Full Text
- View/download PDF
33. Molecular implications of MUC5AC-CD44 axis in colorectal cancer progression and chemoresistance.
- Author
-
Pothuraju R, Rachagani S, Krishn SR, Chaudhary S, Nimmakayala RK, Siddiqui JA, Ganguly K, Lakshmanan I, Cox JL, Mallya K, Kaur S, and Batra SK
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Colorectal Neoplasms drug therapy, Colorectal Neoplasms metabolism, Disease Progression, Fluorouracil administration & dosage, Gene Expression Regulation, Neoplastic, Humans, Hyaluronan Receptors genetics, Mice, Mice, Nude, Mucin 5AC genetics, Oxaliplatin administration & dosage, Prognosis, Survival Rate, Tumor Cells, Cultured, Wnt Signaling Pathway, Xenograft Model Antitumor Assays, beta Catenin genetics, beta Catenin metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Biomarkers, Tumor metabolism, Colorectal Neoplasms pathology, Drug Resistance, Neoplasm, Hyaluronan Receptors metabolism, Mucin 5AC metabolism
- Abstract
Background: Differential expression of mucins has been associated with several cancers including colorectal cancer (CRC). In normal physiological conditions, secretory mucin MUC5AC is not expressed in the colonic mucosa, whereas its aberrant expression is observed during development of colon cancer and its precursor lesions. To date, the molecular mechanism of MUC5AC in CRC progression and drug resistance remains obscure., Methods: MUC5AC expression was determined in colon tissue microarray by immunohistochemistry. A RNA interference and CRISPR/Cas9-mediated system was used to knockdown/knockout the MUC5AC in CRC cell lines to delineate its role in CRC tumorigenesis using in vitro functional assays and in vivo (sub-cutaneous and colon orthotopic) mouse models. Finally, CRC cell lines and xenograft models were used to identify the mechanism of action of MUC5AC., Results: Overexpression of MUC5AC is observed in CRC patient tissues and cell lines. MUC5AC expression resulted in enhanced cell invasion and migration, and decreased apoptosis of CRC cells. MUC5AC interacted with CD44 physically, which was accompanied by the activation of Src signaling. Further, the presence of MUC5AC resulted in enhanced tumorigenesis and appearance of metastatic lesions in orthotopic mouse model. Additionally, up-regulation of MUC5AC resulted in resistance to 5-fluorouracil (5-FU) and oxaliplatin, and its knockout increased sensitivity to these drugs. Finally, we observed that up-regulation of MUC5AC conferred resistance to 5-FU through down-regulation of p53 and its target gene p21 and up-regulation of β-catenin and its target genes CD44 and Lgr5., Conclusion: Our findings suggest that differential expression of secretory mucin MUC5AC results in enhanced tumorigenesis and also confers chemoresistance via CD44/β-catenin/p53/p21 signaling.
- Published
- 2020
- Full Text
- View/download PDF
34. A phase I study of weekly doxorubicin and oral topotecan for patients with relapsed or refractory small cell lung cancer (SCLC): A Fred and Pamela Buffet Cancer Center Clinical Trials Network study.
- Author
-
Ernani V, Jahan R, Smith LM, Marr AS, Kimbrough SE, Kos ME, Tijerina J, Pivovar S, Lakshmanan I, Ketcham M, Rauth S, Mallya K, Nasser MW, Jain M, Kessinger A, Batra SK, and Ganti AK
- Subjects
- Administration, Oral, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Aspartate Aminotransferases blood, Chemical and Drug Induced Liver Injury blood, Chemical and Drug Induced Liver Injury diagnosis, Chemical and Drug Induced Liver Injury epidemiology, Chemical and Drug Induced Liver Injury etiology, Dose-Response Relationship, Drug, Doxorubicin administration & dosage, Drug Administration Schedule, Drug Resistance, Neoplasm, Female, Humans, Lung Neoplasms blood, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Recurrence, Local blood, Neoplasm Recurrence, Local mortality, Neoplasm Recurrence, Local pathology, Progression-Free Survival, Response Evaluation Criteria in Solid Tumors, Severity of Illness Index, Small Cell Lung Carcinoma blood, Small Cell Lung Carcinoma mortality, Small Cell Lung Carcinoma pathology, Thrombocytopenia blood, Thrombocytopenia chemically induced, Thrombocytopenia diagnosis, Thrombocytopenia epidemiology, Topotecan administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Doxorubicin adverse effects, Lung Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Small Cell Lung Carcinoma drug therapy, Topotecan adverse effects
- Abstract
Background: Relapsed/refractory small cell lung cancer (SCLC) has a poor prognosis, with no good options. We evaluated a novel combination of topotecan and doxorubicin, providing sequential topoisomerase I and II inhibition, in this setting., Materials and Methods: Adult patients (>19 years) with relapsed/refractory SCLC, who had received at least one prior chemotherapy regimen were eligible. Patients received escalating doses of oral topotecan on days 1-5 of each three week cycle (maximum - 5 cycles). The dosing cohorts were: 0.85 mg/m
2 , 1.05 mg/m2 , 1.35 mg/m2 , 1.65 mg/m2 and 2.30 mg/m2 . All patients received weekly doxorubicin 20 mg/m2 intravenously starting day 6 of the first cycle and continued weekly for a maximum of 15 weeks. In the absence of pre-specified dose limiting toxicities (DLT), patients were enrolled serially to escalated dose level cohorts., Results: Twenty-two patients were enrolled, of which 20 were evaluable. Median age was 61 years; 74% were male and 95% were Caucasian. Hematologic side effects were the most common adverse events. There were no therapy-related Grade 5 toxicities. Incidence of DLT based on cohorts were: DL2: 1/6 (Grade 4 thrombocytopenia), DL3: 1/6 (AST elevation) and DL4: 2/4 (Grade 4 thrombocytopenia). Response rate was 20% (4/20) and disease control rate (SD + PR) was 36%. The median progression free and overall survival were 3.6 months and 6 months, respectively., Conclusions: The combination of topotecan and doxorubicin was safe and effective in relapsed/refractory SCLC. The maximum tolerated dose of oral topotecan was 1.35 mg/m2 when given concurrently with weekly doxorubicin., Competing Interests: Declaration of Competing Interest This work was supported by the Fred & Pamela Buffett Cancer Center Support Grant (P30CA036727)., (Copyright © 2019. Published by Elsevier Ltd.)- Published
- 2020
- Full Text
- View/download PDF
35. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells.
- Author
-
Vengoji R, Macha MA, Nimmakayala RK, Rachagani S, Siddiqui JA, Mallya K, Gorantla S, Jain M, Ponnusamy MP, Batra SK, and Shonka N
- Subjects
- Afatinib pharmacology, Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, ErbB Receptors genetics, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma metabolism, Humans, Mice, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Proto-Oncogene Proteins c-met metabolism, Signal Transduction drug effects, Temozolomide pharmacology, Xenograft Model Antitumor Assays, Afatinib administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Temozolomide administration & dosage
- Abstract
Background: Glioblastoma (GBM) is an aggressive brain tumor with universal recurrence and poor prognosis. The recurrence is largely driven by chemoradiation resistant cancer stem cells (CSCs). Epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are amplified in ~ 60% and ~ 30% of GBM patients, respectively; however, therapies targeting EGFR have failed to improve disease outcome. EGFRvIII-mediated cross-activation of tyrosine kinase receptor, cMET, regulates GBM CSC maintenance and promote tumor recurrence. Here, we evaluated the efficacy of pan-EGFR inhibitor afatinib and Temozolomide (TMZ) combination on GBM in vitro and in vivo., Methods: We analyzed the effect of afatinib and temozolomide (TMZ) combination on GBM cells U87MG and U251 engineered to express wild type (WT) EGFR, EGFRvIII or EGFRvIII dead kinase, CSCs isolated from U87 and U87EGFRvIII in vitro. The therapeutic utility of the drug combination was investigated on tumor growth and progression using intracranially injected U87EGFRvIII GBM xenografts., Results: Afatinib and TMZ combination synergistically inhibited the proliferation, clonogenic survival, motility, invasion and induced senescence of GBM cells compared to monotherapy. Mechanistically, afatinib decreased U87EGFRvIII GBM cell proliferation and motility/invasion by inhibiting EGFRvIII/AKT, EGFRvIII/JAK2/STAT3, and focal adhesion kinase (FAK) signaling pathways respectively. Interestingly, afatinib specifically inhibited EGFRvIII-cMET crosstalk in CSCs, resulting in decreased expression of Nanog and Oct3/4, and in combination with TMZ significantly decreased their self-renewal property in vitro. More interestingly, afatinib and TMZ combination significantly decreased the xenograft growth and progression compared to single drug alone., Conclusion: Our study demonstrated significant inhibition of GBM tumorigenicity, CSC maintenance in vitro, and delayed tumor growth and progression in vivo by combination of afatinib and TMZ. Our results warrant evaluation of this drug combination in EGFR and EGFRvIII amplified GBM patients.
- Published
- 2019
- Full Text
- View/download PDF
36. A Combination of MUC5AC and CA19-9 Improves the Diagnosis of Pancreatic Cancer: A Multicenter Study.
- Author
-
Kaur S, Smith LM, Patel A, Menning M, Watley DC, Malik SS, Krishn SR, Mallya K, Aithal A, Sasson AR, Johansson SL, Jain M, Singh S, Guha S, Are C, Raimondo M, Hollingsworth MA, Brand RE, and Batra SK
- Subjects
- Adenoma, Islet Cell metabolism, Carcinoma, Pancreatic Ductal diagnosis, Case-Control Studies, Enzyme-Linked Immunosorbent Assay, Humans, Immunohistochemistry, Multivariate Analysis, Pancreatic Neoplasms diagnosis, Pancreatitis, Chronic metabolism, Radioimmunoassay, Sensitivity and Specificity, Biomarkers, Tumor metabolism, CA-19-9 Antigen metabolism, Carcinoma, Pancreatic Ductal metabolism, Mucin 5AC metabolism, Pancreatic Neoplasms metabolism
- Abstract
Objectives: Pancreatic cancer (PC) is a lethal malignancy that lacks specific diagnostic markers. The present study explores the diagnostic potential of the most differentially overexpressed secretory mucin MUC5AC alone and in combination with CA19-9 using multi-center training and validation sets., Methods: The expression of MUC5AC in benign pancreatic pathologies, PC precursor lesions, primary PC tissues and metastatic lesions was evaluated by immunohistochemistry. Circulating MUC5AC levels were measured using sandwich ELISA assay developed in-house, and CA19-9 was measured using radioimmunoassay. A combined training set (n=346) was used to evaluate the diagnostic (n=241) and predictive (n=105, total samples 201 from pre- and post-surgical and chemotherapy set) significance of MUC5AC. Results were further validated with a pre-defined cut-off value using independent sets from the Mayo Clinic (n=94) and the University of Pittsburgh Medical Center (n=321)., Results: Tissue expression analyses indicated the de novo expression of MUC5AC in pancreatic intraepithelial precursor lesions 1A (PanIN1A); the expression was maintained through all stages of progression to invasive adenocarcinoma. The median circulating MUC5AC levels in patients with resectable early-stage PC (EPC) (stage 1/2; 67.2 ng/ml, IQR: 23.9-382.1) and unresectable late-stage PC (LPC) (stage 3/4; 389.7 ng/ml, IQR: 87.7-948.6) were significantly higher compared with (P-value ≤0.0001) benign controls (BC) (7.2 ng/ml, IQR: 0.4-26.5) and (P-value ≤0.0001) chronic pancreatitis (CP) controls (8.4 ng/ml, IQR: 1.5-19.2). In the diagnostic training set (n=241), MUC5AC efficiently differentiated EPC from healthy controls (HC) (83%/80% sensitive (SN)/specific (SP)), BC (67%/87% SN/SP), and CP (83%/77% SN/SP). Independent validation sets from the Mayo Clinic and UPMC confirmed the diagnostic potential of MUC5AC to differentiate EPC from BC (68%/73%; 65%/83%) and CP (68%/79%; 65%/72%). Furthermore, MUC5AC and CA19-9 combination significantly improved (p-value < 0.001) the diagnostic accuracy for differentiating resectable cases from controls., Conclusions: MUC5AC is a valuable diagnostic biomarker, either alone or in combination with CA19-9, to differentiate PC from CP and benign controls.
- Published
- 2017
- Full Text
- View/download PDF
37. MUC4 is negatively regulated through the Wnt/β-catenin pathway via the Notch effector Hath1 in colorectal cancer.
- Author
-
Pai P, Rachagani S, Dhawan P, Sheinin YM, Macha MA, Qazi AK, Chugh S, Ponnusamy MP, Mallya K, Pothuraju R, and Batra SK
- Abstract
MUC4 is a transmembrane mucin lining the normal colonic epithelium. The aberrant/de novo over-expression of MUC4 is well documented in malignancies of the pancreas, ovary and breast. However, studies have reported the loss of MUC4 expression in the majority of colorectal cancers (CRCs). A MUC4 promoter analysis showed the presence of three putative TCF/LEF sites, implying a possible regulation by the Wnt/β-catenin pathway, which has been shown to drive CRC progression. Thus, the objective of our study was to determine whether MUC4 is regulated by β-catenin in CRC. We first knocked down (KD) β-catenin in three CRC cell lines; LS180, HCT-8 and HCT116, which resulted in increased MUC4 transcript and MUC4 protein. Additionally, the overexpression of stabilized mutant β-catenin in LS180 and HCT-8 resulted in a decrease in MUC4 expression. Immunohistochemistry (IHC) of mouse colon tissue harboring tubular adenomas and high grade dysplasia showed dramatically reduced Muc4 in lesions relative to adjacent normal tissue, with increased cytosolic/nuclear β-catenin. Luciferase assays with the complete MUC4 promoter construct p3778 showed increased MUC4 promoter luciferase activity in the absence of β-catenin (KD). Mutation of all three putative TCF/LEF sites showed that MUC4 promoter luciferase activity was increased relative to the un-mutated promoter. Interestingly, it was observed that MUC4 expressing CRC cell lines also expressed high levels of Hath1, a transcription factor repressed by both active Wnt/β-catenin and Notch signaling. The KD of β-catenin and/or treatment with a Notch γ-secretase inhibitor, Dibenzazepine (DBZ) resulted in increased Hath1 and MUC4 in LS180, HCT-8 and HCT116. Furthermore, overexpression of Hath1 in HCT-8 and LS180 caused increased MUC4 transcript and MUC4 protein. Taken together, our results indicate that the Wnt/β-catenin pathway suppresses the Notch pathway effector Hath1, resulting in reduced MUC4 in CRC.
- Published
- 2016
- Full Text
- View/download PDF
38. Tumour exosome integrins determine organotropic metastasis.
- Author
-
Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, and Lyden D
- Subjects
- Animals, Biomarkers metabolism, Brain cytology, Cell Line, Tumor, Endothelial Cells cytology, Endothelial Cells metabolism, Epithelial Cells cytology, Epithelial Cells metabolism, Female, Fibroblasts cytology, Fibroblasts metabolism, Genes, src, Humans, Integrin alpha6beta1 metabolism, Integrin alpha6beta4 antagonists & inhibitors, Integrin alpha6beta4 metabolism, Integrin beta Chains metabolism, Integrin beta4 metabolism, Integrins antagonists & inhibitors, Kupffer Cells cytology, Kupffer Cells metabolism, Liver cytology, Lung cytology, Mice, Mice, Inbred C57BL, Organ Specificity, Phosphorylation, Receptors, Vitronectin antagonists & inhibitors, Receptors, Vitronectin metabolism, S100 Proteins genetics, Brain metabolism, Exosomes metabolism, Integrins metabolism, Liver metabolism, Lung metabolism, Neoplasm Metastasis pathology, Neoplasm Metastasis prevention & control, Tropism
- Abstract
Ever since Stephen Paget's 1889 hypothesis, metastatic organotropism has remained one of cancer's greatest mysteries. Here we demonstrate that exosomes from mouse and human lung-, liver- and brain-tropic tumour cells fuse preferentially with resident cells at their predicted destination, namely lung fibroblasts and epithelial cells, liver Kupffer cells and brain endothelial cells. We show that tumour-derived exosomes uptaken by organ-specific cells prepare the pre-metastatic niche. Treatment with exosomes from lung-tropic models redirected the metastasis of bone-tropic tumour cells. Exosome proteomics revealed distinct integrin expression patterns, in which the exosomal integrins α6β4 and α6β1 were associated with lung metastasis, while exosomal integrin αvβ5 was linked to liver metastasis. Targeting the integrins α6β4 and αvβ5 decreased exosome uptake, as well as lung and liver metastasis, respectively. We demonstrate that exosome integrin uptake by resident cells activates Src phosphorylation and pro-inflammatory S100 gene expression. Finally, our clinical data indicate that exosomal integrins could be used to predict organ-specific metastasis.
- Published
- 2015
- Full Text
- View/download PDF
39. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver.
- Author
-
Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, García-Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz RE, Matei I, Peinado H, Stanger BZ, Bromberg J, and Lyden D
- Subjects
- Animals, Base Sequence, Bone Marrow Cells immunology, Cell Line, Tumor, Cell Movement, Female, Fibronectins biosynthesis, Gene Expression Regulation, Neoplastic, Hepatic Stellate Cells pathology, Humans, Liver cytology, Liver pathology, Liver Neoplasms genetics, Liver Neoplasms secondary, Macrophages immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Precancerous Conditions pathology, RNA Interference, RNA, Small Interfering, Sequence Analysis, RNA, Signal Transduction, Transforming Growth Factor beta metabolism, Carcinoma, Pancreatic Ductal pathology, Exosomes metabolism, Liver Neoplasms pathology, Macrophage Migration-Inhibitory Factors biosynthesis, Pancreatic Neoplasms pathology
- Abstract
Pancreatic ductal adenocarcinomas (PDACs) are highly metastatic with poor prognosis, mainly due to delayed detection. We hypothesized that intercellular communication is critical for metastatic progression. Here, we show that PDAC-derived exosomes induce liver pre-metastatic niche formation in naive mice and consequently increase liver metastatic burden. Uptake of PDAC-derived exosomes by Kupffer cells caused transforming growth factor β secretion and upregulation of fibronectin production by hepatic stellate cells. This fibrotic microenvironment enhanced recruitment of bone marrow-derived macrophages. We found that macrophage migration inhibitory factor (MIF) was highly expressed in PDAC-derived exosomes, and its blockade prevented liver pre-metastatic niche formation and metastasis. Compared with patients whose pancreatic tumours did not progress, MIF was markedly higher in exosomes from stage I PDAC patients who later developed liver metastasis. These findings suggest that exosomal MIF primes the liver for metastasis and may be a prognostic marker for the development of PDAC liver metastasis.
- Published
- 2015
- Full Text
- View/download PDF
40. MUC4-mediated regulation of acute phase protein lipocalin 2 through HER2/AKT/NF-κB signaling in pancreatic cancer.
- Author
-
Kaur S, Sharma N, Krishn SR, Lakshmanan I, Rachagani S, Baine MJ, Smith LM, Lele SM, Sasson AR, Guha S, Mallya K, Anderson JM, Hollingsworth MA, and Batra SK
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Gene Knockdown Techniques, Humans, Immunoblotting, Immunohistochemistry, Lipocalin-2, Mice, Microscopy, Confocal, Microscopy, Fluorescence, NF-kappa B metabolism, Oncogene Proteins metabolism, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins c-akt metabolism, Real-Time Polymerase Chain Reaction, Receptor, ErbB-2 metabolism, Acute-Phase Proteins metabolism, Biomarkers, Tumor analysis, Lipocalins metabolism, Mucin-4 metabolism, Pancreatic Neoplasms metabolism, Proto-Oncogene Proteins metabolism, Signal Transduction physiology
- Abstract
Purpose: MUC4 shows aberrant expression in early pancreatic lesions and a high specificity for pancreatic cancer. It thus has a high potential to be a sensitive and specific biomarker. Unfortunately, its low serum level limits its diagnostic/prognostic potential. We here report that a multifaceted acute phase protein lipocalin 2, regulated by MUC4, could be a potential diagnostic/prognostic marker for pancreatic cancer. Experimental Designs and, Results: Overexpression/knockdown, luciferase reporter and molecular inhibition studies revealed that MUC4 regulates lipocalin 2 by stabilizing HER2 and stimulating AKT, which results in the activation of NF-κB. Immunohistochemical analyses of lipocalin 2 and MUC4 showed a significant positive correlation between MUC4 and lipocalin 2 in primary, metastatic tissues (Spearman correlation coefficient 0.71, P = 0.002) from rapid autopsy tissue sample from patients with pancreatic cancer as well as in serum and tissue samples from spontaneous KRASG(12)D mouse pancreatic cancer model (Spearman correlation coefficient 0.98, P < 0.05). Lipocalin 2 levels increased progressively with disease advancement (344.2 ± 22.8 ng/mL for 10 weeks to 3067.2 ± 572.6 for 50 weeks; P < 0.0001). In human pancreatic cancer cases, significantly elevated levels of lipocalin 2 were observed in patients with pancreatic cancer (148 ± 13.18 ng/mL) in comparison with controls (73.27 ± 4.9 ng/mL, P = 0.014). Analyses of pre- and postchemotherapy patients showed higher lipocalin 2 levels in prechemotherapy patients [121.7 ng/mL; 95% confidence interval (CI), 98.1-150.9] in comparison with the postchemotherapy (92.6 ng/mL; 95% CI, 76.7-111.6; P = 0.06) group., Conclusions: This study delineates the association and the downstream mechanisms of MUC4-regulated elevation of lipocalin-2 (via HER2/AKT/NF-κB) and its clinical significance for prognosis of pancreatic cancer., (©2013 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
41. Novel pancreatic cancer cell lines derived from genetically engineered mouse models of spontaneous pancreatic adenocarcinoma: applications in diagnosis and therapy.
- Author
-
Torres MP, Rachagani S, Souchek JJ, Mallya K, Johansson SL, and Batra SK
- Subjects
- Adenocarcinoma diagnosis, Adenocarcinoma therapy, Animals, Base Sequence, Blotting, Western, Cell Line, Tumor, DNA Primers, Mice, Microscopy, Confocal, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms therapy, Real-Time Polymerase Chain Reaction, Adenocarcinoma pathology, Pancreatic Neoplasms pathology
- Abstract
Pancreatic cancer (PC) remains one of the most lethal human malignancies with poor prognosis. Despite all advances in preclinical research, there have not been significant translation of novel therapies into the clinics. The development of genetically engineered mouse (GEM) models that produce spontaneous pancreatic adenocarcinoma (PDAC) have increased our understanding of the pathogenesis of the disease. Although these PDAC mouse models are ideal for studying potential therapies and specific genetic mutations, there is a need for developing syngeneic cell lines from these models. In this study, we describe the successful establishment and characterization of three cell lines derived from two (PDAC) mouse models. The cell line UN-KC-6141 was derived from a pancreatic tumor of a Kras(G12D);Pdx1-Cre (KC) mouse at 50 weeks of age, whereas UN-KPC-960 and UN-KPC-961 cell lines were derived from pancreatic tumors of Kras(G12D);Trp53(R172H);Pdx1-Cre (KPC) mice at 17 weeks of age. The cancer mutations of these parent mice carried over to the daughter cell lines (i.e. Kras(G12D) mutation was observed in all three cell lines while Trp53 mutation was observed only in KPC cell lines). The cell lines showed typical cobblestone epithelial morphology in culture, and unlike the previously established mouse PDAC cell line Panc02, expressed the ductal marker CK19. Furthermore, these cell lines expressed the epithelial-mesenchymal markers E-cadherin and N-cadherin, and also, Muc1 and Muc4 mucins. In addition, these cell lines were resistant to the chemotherapeutic drug Gemcitabine. Their implantation in vivo produced subcutaneous as well as tumors in the pancreas (orthotopic). The genetic mutations in these cell lines mimic the genetic compendium of human PDAC, which make them valuable models with a high potential of translational relevance for examining diagnostic markers and therapeutic drugs.
- Published
- 2013
- Full Text
- View/download PDF
42. Neutrophil gelatinase-associated lipocalin, macrophage inhibitory cytokine 1, and carbohydrate antigen 19-9 in pancreatic juice: pathobiologic implications in diagnosing benign and malignant disease of the pancreas.
- Author
-
Kaur S, Baine MJ, Guha S, Ochi N, Chakraborty S, Mallya K, Thomas C, Crook J, Wallace MB, Woodward TA, Jain M, Singh S, Sasson AR, Skinner V, Raimondo M, and Batra SK
- Subjects
- Aged, Biomarkers metabolism, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 metabolism, Diagnosis, Differential, Enzyme-Linked Immunosorbent Assay, Female, Humans, Lipocalin-2, Male, Middle Aged, Pancreas metabolism, Pancreas pathology, Pancreatic Neoplasms complications, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms metabolism, Pancreatitis, Chronic complications, Pancreatitis, Chronic diagnosis, Pancreatitis, Chronic metabolism, Prospective Studies, Radioimmunoassay, Sensitivity and Specificity, Acute-Phase Proteins metabolism, CA-19-9 Antigen metabolism, Growth Differentiation Factor 15 metabolism, Lipocalins metabolism, Pancreatic Juice metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Objective: Pancreatic diseases pose significant diagnostic challenge as signs and symptoms often overlap. We investigated the potential of pancreatic juice neutrophil gelatinase-associated lipocalin, macrophage inhibitory cytokine 1 (MIC-1), and carbohydrate antigen 19-9 (CA19-9) to aid in the diagnosis of patients with symptoms suggestive of pancreatic diseases., Methods: A total of 105 chronic pancreatitis (CP), pancreatic cancer (PC), and nonpancreatic nonhealthy (patients with symptoms mimicking pancreatic disease but found to be free of any pancreatic disease) patients underwent endoscopic pancreatic juice collection after secretin stimulation. Neutrophil gelatinase-associated lipocalin and MIC-1 levels were measured by enzyme-linked immunosorbent assay, whereas CA19-9 was measured by radioimmunoassay., Results: Neutrophil gelatinase-associated lipocalin, MIC-1, and CA19-9 were significantly elevated in the pancreatic juice of patients with CP and patients with PC as compared with nonpancreatic nonhealthy controls (P ≤ 0.034). Neutrophil gelatinase-associated lipocalin seemed most promising in differentiating diseased versus nondiseased pancreata (areas under the curve, 0.88-0.91), whereas MIC-1 was found to be higher in patients with PC than in patients with CP (P = 0.043). Interestingly, MIC-1 levels in diabetic patients with PC were higher than in nondiabetic patients with PC (P = 0.030) and diabetic patients with CP (P = 0.087). Carbohydrate antigen 19-9 showed the least ability to distinguish patient groups (areas under the curve, 0.61-0.76)., Conclusions: Pancreatic juice neutrophil gelatinase-associated lipocalin shows potential utility in establishing pancreatic etiology in the context of nonspecific symptoms, whereas MIC-1 may aid in differentiating PC from CP.
- Published
- 2013
- Full Text
- View/download PDF
43. Quantitative real-time PCR expression analysis of peripheral blood mononuclear cells in pancreatic cancer patients.
- Author
-
Baine MJ, Mallya K, and Batra SK
- Subjects
- Cell Separation methods, Humans, Specimen Handling, Leukocytes, Mononuclear metabolism, Pancreatic Neoplasms genetics, Real-Time Polymerase Chain Reaction methods
- Abstract
The ability of peripheral blood mononuclear cells (PBMCs) to act as a surrogate window into the presence and physiologic effects of pancreatic cancer is becoming increasingly apparent. In this chapter, we describe the techniques for isolation, lysis, RNA extraction, cDNA synthesis, and Q-RT PCR analysis of PBMCs as well as reasonable alternatives and the advantages and disadvantages of each. We further discuss the noteworthy considerations necessary for successful isolation and conversion of the high-quality PBMC RNA required to acquire interpretable and reproducible results for PBMC genetic expression analysis.
- Published
- 2013
- Full Text
- View/download PDF
44. Potentials of plasma NGAL and MIC-1 as biomarker(s) in the diagnosis of lethal pancreatic cancer.
- Author
-
Kaur S, Chakraborty S, Baine MJ, Mallya K, Smith LM, Sasson A, Brand R, Guha S, Jain M, Wittel U, Singh SK, and Batra SK
- Subjects
- Analysis of Variance, Body Mass Index, CA-19-9 Antigen blood, Diagnosis, Differential, Humans, Lipocalin-2, Logistic Models, Pancreatic Neoplasms blood, Pancreatitis, Chronic diagnosis, Radioimmunoassay, Retrospective Studies, Sensitivity and Specificity, Acute-Phase Proteins, Biomarkers blood, Growth Differentiation Factor 15 blood, Lipocalins blood, Pancreatic Neoplasms diagnosis, Proto-Oncogene Proteins blood
- Abstract
Pancreatic cancer (PC) is lethal malignancy with very high mortality rate. Absence of sensitive and specific marker(s) is one of the major factors for poor prognosis of PC patients. In pilot studies using small set of patients, secreted acute phase proteins neutrophil gelatinase associated lipocalin (NGAL) and TGF-β family member macrophage inhibitory cytokine-1 (MIC-1) are proposed as most potential biomarkers specifically elevated in the blood of PC patients. However, their performance as diagnostic markers for PC, particularly in pre-treatment patients, remains unknown. In order to evaluate the diagnostic efficacy of NGAL and MIC-1, their levels were measured in plasma samples from patients with pre-treatment PC patients (n = 91) and compared it with those in healthy control (HC) individuals (n = 24) and patients with chronic pancreatitis (CP, n = 23). The diagnostic performance of these two proteins was further compared with that of CA19-9, a tumor marker commonly used to follow PC progression. The levels of all three biomarkers were significantly higher in PC compared to HCs. The mean (± standard deviation, SD) plasma NGAL, CA19-9 and MIC-1 levels in PC patients was 111.1 ng/mL (2.2), 219.2 U/mL (7.8) and 4.5 ng/mL (4.1), respectively. In comparing resectable PC to healthy patients, all three biomarkers were found to have comparable sensitivities (between 64%-81%) but CA19-9 and NGAL had a higher specificity (92% and 88%, respectively). For distinguishing resectable PC from CP patients, CA19-9 and MIC-1 were most specific (74% and 78% respectively). CA19-9 at an optimal cut-off of 54.1 U/ml is highly specific in differentiating resectable (stage 1/2) pancreatic cancer patients from controls in comparison to its clinical cut-off (37.1 U/ml). Notably, the addition of MIC-1 to CA19-9 significantly improved the ability to distinguish resectable PC cases from CP (p = 0.029). Overall, MIC-1 in combination with CA19-9 improved the diagnostic accuracy of differentiating PC from CP and HCs.
- Published
- 2013
- Full Text
- View/download PDF
45. Transcriptional profiling of peripheral blood mononuclear cells in pancreatic cancer patients identifies novel genes with potential diagnostic utility.
- Author
-
Baine MJ, Chakraborty S, Smith LM, Mallya K, Sasson AR, Brand RE, and Batra SK
- Subjects
- Case-Control Studies, Female, Humans, Male, Middle Aged, Pancreatic Neoplasms pathology, Reproducibility of Results, Sensitivity and Specificity, Leukocytes, Mononuclear metabolism, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms genetics, Transcription, Genetic genetics, Transcriptome
- Abstract
Background: It is well known that many malignancies, including pancreatic cancer (PC), possess the ability to evade the immune system by indirectly downregulating the mononuclear cell machinery necessary to launch an effective immune response. This knowledge, in conjunction with the fact that the trancriptome of peripheral blood mononuclear cells has been shown to be altered in the context of many diseases, including renal cell carcinoma, lead us to study if any such alteration in gene expression exists in PC as it may have diagnostic utility., Methods and Findings: PBMC samples from 26 PC patients and 33 matched healthy controls were analyzed by whole genome cDNA microarray. Three hundred eighty-three genes were found to be significantly different between PC and healthy controls, with 65 having at least a 1.5 fold change in expression. Pathway analysis revealed that many of these genes fell into pathways responsible for hematopoietic differentiation, cytokine signaling, and natural killer (NK) cell and CD8+ T-cell cytotoxic response. Unsupervised hierarchical clustering analysis identified an eight-gene predictor set, consisting of SSBP2, Ube2b-rs1, CA5B, F5, TBC1D8, ANXA3, ARG1, and ADAMTS20, that could distinguish PC patients from healthy controls with an accuracy of 79% in a blinded subset of samples from treatment naïve patients, giving a sensitivity of 83% and a specificity of 75%., Conclusions: In summary, we report the first in-depth comparison of global gene expression profiles of PBMCs between PC patients and healthy controls. We have also identified a gene predictor set that can potentially be developed further for use in diagnostic algorithms in PC. Future directions of this research should include analysis of PBMC expression profiles in patients with chronic pancreatitis as well as increasing the number of early-stage patients to assess the utility of PBMCs in the early diagnosis of PC.
- Published
- 2011
- Full Text
- View/download PDF
46. Differential gene expression analysis of peripheral blood mononuclear cells reveals novel test for early detection of pancreatic cancer.
- Author
-
Baine MJ, Menning M, Smith LM, Mallya K, Kaur S, Rachagani S, Chakraborty S, Sasson AR, Brand RE, and Batra SK
- Subjects
- Aged, Area Under Curve, CA-19-9 Antigen genetics, CA-19-9 Antigen metabolism, Carbonic Anhydrase V genetics, Carbonic Anhydrase V metabolism, Case-Control Studies, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Factor V genetics, Factor V metabolism, Female, Growth Differentiation Factor 15 genetics, Growth Differentiation Factor 15 metabolism, Humans, Male, Middle Aged, Molecular Diagnostic Techniques, Multiplex Polymerase Chain Reaction, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms genetics, ROC Curve, Real-Time Polymerase Chain Reaction, Transcription, Genetic, Transcriptome, Early Detection of Cancer, Leukocytes, Mononuclear metabolism, Pancreatic Neoplasms blood
- Abstract
Background: We sought to validate global microarray results indicating the differential expression of 383 genes in Peripheral Blood Mononuclear Cells (PBMCs) from patients with pancreatic cancer (PC) and to further evaluate their PC diagnostic potential., Methods and Materials: In total, 177 patients were recruited (47 healthy controls (HC), 35 chronic pancreatitis (CP) patients, and 95 PC patients). PBMC expressions of six genes from our previous study (ANXA3, ARG1, CA5B, F5, SSBP2, and TBC1D8) along with four new genes (MIC1, NGAL, MUC1, and MUC16) were analyzed using multiplex Q-RT PCR., Results: Differential expressions of 5 of the 6 genes previously identified by PBMC microarray were validated in this study. Multivariate models for PBMC gene expression were attempted to determine if any combination was diagnostically superior to CA19-9 alone. We found that addition of PBMC CA5B, F5, SSBP2, and MIC1 expression levels to CA19-9 significantly improved CA19-9's diagnostic abilities when comparing resectable PC to CP patients (p=0.023)., Conclusions: Results of our previous study were validated, indicating reproducibility of PC-associated PBMC expression profiling. We identified a score-based model that can differentiate resectable PC from CP better than CA19-9, potentiating that PBMC differential expression analysis may offer a novel tool for early PC diagnosis.
- Published
- 2011
- Full Text
- View/download PDF
47. The canonical Wnt pathway regulates retinal stem cells/progenitors in concert with Notch signaling.
- Author
-
Das AV, Bhattacharya S, Zhao X, Hegde G, Mallya K, Eudy JD, and Ahmad I
- Subjects
- Animals, Cell Differentiation physiology, Cell Proliferation, Chick Embryo, Female, Lymphoid Enhancer-Binding Factor 1 genetics, Lymphoid Enhancer-Binding Factor 1 metabolism, Oligonucleotide Array Sequence Analysis, Pregnancy, Rats, Rats, Sprague-Dawley, Receptors, Notch genetics, Retina embryology, Stem Cells cytology, Wnt Proteins genetics, Receptors, Notch metabolism, Retina cytology, Retina metabolism, Signal Transduction physiology, Stem Cells physiology, Wnt Proteins metabolism
- Abstract
The canonical Wnt pathway is known to influence multiple developmental events such as patterning, cell proliferation and cell specification. Recent studies have provided evidence of the involvement of the canonical Wnt pathway in the emergence and development of the optic neuroepithelium and its derivatives, particularly the retina. However, the mechanism of its action during retinal development remains rather obscure. Here, we demonstrate that (in agreement with observations in the blood, intestine, and skin) the canonical Wnt pathway influences retinal development by maintaining stem cells/progenitors. For example, the activation of this pathway keeps the early retinal stem cells/progenitors proliferating and uncommitted, while its attenuation facilitates their differentiation into retinal ganglion cells in vitro and in vivo. In addition, we demonstrate that Wnt signaling acts in concert with Notch signaling during retinal histogenesis, where the latter calibrates the influence of the former on the differentiation status of retinal stem cells/progenitors by regulating Lef1 and sFRP2., (2008 S. Karger AG, Basel.)
- Published
- 2008
- Full Text
- View/download PDF
48. SWI/SNF chromatin remodeling ATPase Brm regulates the differentiation of early retinal stem cells/progenitors by influencing Brn3b expression and Notch signaling.
- Author
-
Das AV, James J, Bhattacharya S, Imbalzano AN, Antony ML, Hegde G, Zhao X, Mallya K, Ahmad F, Knudsen E, and Ahmad I
- Subjects
- Adenosine Triphosphatases genetics, Animals, Cell Cycle Proteins genetics, Cell Differentiation, Cell Lineage, Cell Proliferation, Chromatin metabolism, DNA Helicases, Models, Biological, Nuclear Proteins, Rats, Rats, Sprague-Dawley, Retina cytology, Signal Transduction, Stem Cells cytology, Transcription, Genetic, Adenosine Triphosphatases physiology, Cell Cycle Proteins physiology, Gene Expression Regulation, Gene Expression Regulation, Developmental, Receptor, Notch1 biosynthesis, Retina embryology, Transcription Factor Brn-3B biosynthesis, Transcription Factors metabolism
- Abstract
Based on a variety of approaches, evidence suggests that different cell types in the vertebrate retina are generated by multipotential progenitors in response to interactions between cell intrinsic and cell extrinsic factors. The identity of some of the cellular determinants that mediate such interactions has emerged, shedding light on mechanisms underlying cell differentiation. For example, we know now that Notch signaling mediates the influence of the microenvironment on states of commitment of the progenitors by activating transcriptional repressors. Cell intrinsic factors such as the proneural basic helix-loop-helix and homeodomain transcription factors regulate a network of genes necessary for cell differentiation and maturation. What is missing from this picture is the role of developmental chromatin remodeling in coordinating the expression of disparate classes of genes for the differentiation of retinal progenitors. Here we describe the role of Brm, an ATPase in the SWI/SNF chromatin remodeling complex, in the differentiation of retinal progenitors into retinal ganglion cells. Using the perturbation of expression and function analyses, we demonstrate that Brm promotes retinal ganglion cell differentiation by facilitating the expression and function of a key regulator of retinal ganglion cells, Brn3b, and the inhibition of Notch signaling. In addition, we demonstrate that Brm promotes cell cycle exit during retinal ganglion cell differentiation. Together, our results suggest that Brm represents one of the nexus where diverse information of cell differentiation is integrated during cell differentiation.
- Published
- 2007
- Full Text
- View/download PDF
49. Maintenance of retinal stem cells by Abcg2 is regulated by notch signaling.
- Author
-
Bhattacharya S, Das A, Mallya K, and Ahmad I
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters genetics, Animals, Cell Differentiation, Cells, Cultured cytology, Cells, Cultured metabolism, Electrophoretic Mobility Shift Assay, Female, Gene Expression Regulation, Metabolic Networks and Pathways, RNA Interference, Rats, Rats, Sprague-Dawley, Retina metabolism, Stem Cells metabolism, ATP-Binding Cassette Transporters metabolism, Receptors, Notch metabolism, Retina cytology, Signal Transduction, Stem Cells cytology
- Abstract
ABCG2 belongs to the ATP-binding cassette superfamily of transmembrane proteins and is ubiquitously expressed in stem cells including those in the developing nervous system. The ability of ABCG2 to preferentially exclude DNA-intercalating dyes is regarded to be the basis for the enrichment of stem cells or progenitors as dye(low) side population (SP) cells. However, the role of ABCG2 in neural stem cells remains speculative and poorly understood. Here, we demonstrate using retinal stem cells, that ABCG2 is the molecular determinant of SP cell phenotype of neural stem cells and plays an important role in their maintenance. Overexpression of ABCG2 prevents the SP cell phenotype and adversely affects the lineage commitment of retinal stem cells. By contrast, targeted attenuation of ABCG2 depletes retinal SP cells and promotes their differentiation along pan neural and retinal lineages. In addition, we demonstrate for the first time that ABCG2 is a target of Notch signaling, and as such, constitutes one of the genes in the regulatory network of Notch signaling, involved in the maintenance of stem cells.
- Published
- 2007
- Full Text
- View/download PDF
50. The biomimetic [Cr3O(O2CCH2CH3)6(H2O)3]+ decreases plasma cholesterol and triglycerides in rats: towards chromium-containing therapeutics.
- Author
-
Sun Y, Mallya K, Ramirez J, and Vincent JB
- Subjects
- Animals, Insulin blood, Male, Molecular Mimicry, Rats, Rats, Sprague-Dawley, Cholesterol blood, Hypolipidemic Agents pharmacology, Organometallic Compounds pharmacology, Triglycerides blood
- Abstract
The in vivo effects of administration of the synthetic, functional biomimetic [Cr3O(O2CCH2CH3)6(H2O)3]+ 1 and a naturally occurring, biologically active form of chromium, low-molecular-weight chromium-binding substance (LMWCr), to rats are described. Given that the complexes are proposed to function by interacting with insulin receptor, trapping it in its active conformation, in contrast to current chromium-containing nutrition supplements, which only serve as sources of absorbable chromium, changes in lipid and carbohydrate metabolism would be expected. After 12 weeks administration (20 micrograms/kg body mass), compound 1 results in 40% lower levels of blood plasma LDL cholesterol, 33% lower levels of total cholesterol, and significantly lower HDL cholesterol and triglyceride; these results are in stark contrast to those of administration of other forms of Cr(III) to rats, which have no effect on these parameters. LMWCr, in contrast to 1, has no effect as it probably is degraded in vivo or excreted. These results are interpreted in terms of the mechanism of chromium action in response to insulin and the activation of insulin receptor, and the potential for the rational design of chromium-containing therapeutics is discussed.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.