434 results on '"Surinder K Batra"'
Search Results
2. Rethinking the chemokine cascade in brain metastasis: Preventive and therapeutic implications
- Author
-
Shailendra Kumar Maurya, Parvez Khan, Asad Ur Rehman, Ranjana K. Kanchan, Naveenkumar Perumal, Sidharth Mahapatra, Hitendra S. Chand, Juan A. Santamaria-Barria, Surinder K. Batra, and Mohd Wasim Nasser
- Subjects
Cancer Research ,Brain Neoplasms ,Neoplastic Stem Cells ,Tumor Microenvironment ,Humans ,Brain ,Chemokines ,Neoplasm Metastasis - Abstract
Brain metastasis (BrM) is one of the major causes of death in cancer patients and is associated with an estimated 10-40 % of total cancer cases. The survival rate of brain metastatic patients has not improved due to intratumor heterogeneity, the survival adaptations of brain homing metastatic cells, and the lack of understanding of underlying molecular mechanisms that limit the availability of effective therapies. The heterogeneous population of immune cells and tumor-initiating cells or cancer stem cells in the tumor microenvironment (TME) release various factors, such as chemokines that upon binding to their cognate receptors enhance tumor growth at primary sites and help tumor cells metastasize to the brain. Furthermore, brain metastatic sites have unique heterogeneous microenvironment that fuels cancer cells in establishing BrM. This review explores the crosstalk of chemokines with the heterogeneous TME during the progression of BrM and recognizes potential therapeutic approaches. We also discuss and summarize different targeted, immunotherapeutic, chemotherapeutic, and combinatorial strategies (with chemo-/immune- or targeted-therapies) to attenuate chemokines mediated BrM.
- Published
- 2022
3. Cytokines chattering in pancreatic ductal adenocarcinoma tumor microenvironment
- Author
-
Rakesh Bhatia, Namita Bhyravbhatla, Andrew Kisling, Xiaoqi Li, Surinder K. Batra, and Sushil Kumar
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Epithelial-Mesenchymal Transition ,Carcinogenesis ,Tumor Microenvironment ,Humans ,Cytokines ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME) consists of multiple cell types interspersed by dense fibrous stroma. These cells communicate through low molecular weight signaling molecules called cytokines. The cytokines, through their receptors, facilitate PDAC initiation, progression, metastasis, and distant colonization of malignant cells. These signaling mediators secreted from tumor-associated macrophages, and cancer-associated fibroblasts in conjunction with oncogenic Kras mutation initiate acinar to ductal metaplasia (ADM), resulting in the appearance of early preneoplastic lesions. Further, M1- and M2-polarized macrophages provide proinflammatory conditions and promote deposition of extracellular matrix, whereas myofibroblasts and T-lymphocytes, such as Th17 and T-regulatory cells, create a fibroinflammatory and immunosuppressive environment with a significantly reduced cytotoxic T-cell population. During PDAC progression, cytokines regulate the expression of various oncogenic regulators such as NFκB, c-myc, growth factor receptors, and mucins resulting in the formation of high-grade PanIN lesions, epithelial to mesenchymal transition, invasion, and extravasation of malignant cells, and metastasis. During metastasis, PDAC cells colonize at the premetastatic niche created in the liver, and lung, an organotropic function primarily executed by cytokines in circulation or loaded in the exosomes from the primary tumor cells. The indispensable contribution of these cytokines at every stage of PDAC tumorigenesis makes them exciting candidates in combination with immune-, chemo- and targeted radiation therapy.
- Published
- 2022
4. Chemokine-mucinome interplay in shaping the heterogeneous tumor microenvironment of pancreatic cancer
- Author
-
Koelina, Ganguly, Ashu, Shah, Pranita, Atri, Sanchita, Rauth, Moorthy P, Ponnusamy, Sushil, Kumar, and Surinder K, Batra
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Cancer-Associated Fibroblasts ,Tumor Microenvironment ,Mucins ,Humans ,Chemokines - Abstract
Pancreatic cancer (PC) is exemplified by a complex immune-suppressive, fibrotic tumor microenvironment (TME), and aberrant expression of mucins. The constant crosstalk between cancer cells, cancer-associated fibroblasts (CAFs), and the immune cells mediated by the soluble factors and inflammatory mediators including cytokines, chemokines, reactive oxygen species (ROS) promote the dynamic temporal switch towards an immune-escape phenotype in the neoplastic cells and its microenvironment that bolsters disease progression. Chemokines have been studied in PC pathogenesis, albeit poorly in the context of mucins, tumor glycocalyx, and TME heterogeneity (CAFs and immune cells). With correlative analysis from PC patients' transcriptome data, support from available literature, and scientific arguments-based speculative extrapolations in terms of disease pathogenesis, we have summarized in this review a comprehensive understanding of chemokine-mucinome interplay during stromal modulation and immune-suppression in PC. Future studies should focus on deciphering the complexities of chemokine-mediated control of glycocalyx maturation, immune infiltration, and CAF-associated immune suppression. Knowledge extracted from such studies will be beneficial to mechanistically correlate the mucin-chemokine abundance in serum versus pancreatic tumors of patients, which may aid in prognostication and stratification of PC patients for immunotherapy.
- Published
- 2022
5. Muc16 depletion diminishes KRAS-induced tumorigenesis and metastasis by altering tumor microenvironment factors in pancreatic ductal adenocarcinoma
- Author
-
Imayavaramban Lakshmanan, Saravanakumar Marimuthu, Sanjib Chaudhary, Parthasarathy Seshacharyulu, Satyanarayana Rachagani, Sakthivel Muniyan, Ramakanth Chirravuri-Venkata, Pranita Atri, Sanchita Rauth, Rama Krishna Nimmakayala, Jawed Akhtar Siddiqui, Shailendra K. Gautam, Ashu Shah, Gopalakrishnan Natarajan, Seema Parte, Namita Bhyravbhatla, Kavita Mallya, Dhanya Haridas, Geoffrey A. Talmon, Lynette M. Smith, Sushil Kumar, Apar Kishor Ganti, Maneesh Jain, Moorthy P. Ponnusamy, and Surinder K. Batra
- Subjects
Pancreatic Neoplasms ,Proto-Oncogene Proteins p21(ras) ,Mice ,Cancer Research ,Carcinogenesis ,Mucins ,Tumor Microenvironment ,Genetics ,Animals ,Molecular Biology ,Article ,Carcinoma, Pancreatic Ductal - 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 Trp53(R172H) mutations remains unknown. Deletion of Muc16 with activating mutations Kras(G12D/+) and Trp53(R172H/+) in mice significantly decreased progression and prolonged overall survival in Kras(G12D/+); Trp53(R172H/+); Pdx-1-Cre; Muc16(−/−) (KPCM) and Kras(G12D/+); Pdx-1-Cre; Muc16(−/−) (KCM), as compared to Kras(G12D/+); Trp53(R172H/+); Pdx-1-Cre (KPC) and Kras(G12D/+); 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 than 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.
- Published
- 2022
6. Epigenetic landscape of small cell lung cancer: small image of a giant recalcitrant disease
- Author
-
Mohd W. Nasser, Imayavaramban Lakshmanan, Surinder K. Batra, Jawed A. Siddiqui, Ravi Salgia, Maneesh Jain, Shailendra Kumar Maurya, Apar Kishor Ganti, and Parvez Khan
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Disease ,medicine.disease_cause ,Article ,Epigenesis, Genetic ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Histone methylation ,Humans ,Medicine ,Epigenetics ,Lung cancer ,neoplasms ,business.industry ,Cancer ,DNA Methylation ,medicine.disease ,Small Cell Lung Carcinoma ,humanities ,respiratory tract diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,Immunotherapy ,business ,Carcinogenesis - Abstract
Small cell lung cancer (SCLC) is a particular subtype of lung cancer with high mortality. Recent advances in understanding SCLC genomics and breakthroughs of immunotherapy have substantially expanded existing knowledge and treatment modalities. However, challenges associated with SCLC remain enigmatic and elusive. Most of the conventional drug discovery approaches targeting altered signaling pathways in SCLC end up in the ‘grave-yard of drug discovery’, which mandates exploring novel approaches beyond inhibiting cell signaling pathways. Epigenetic modifications have long been documented as the key contributors to the tumorigenesis of almost all types of cancer, including SCLC. The last decade witnessed an exponential increase in our understanding of epigenetic modifications for SCLC. The present review highlights the central role of epigenetic regulations in acquiring neoplastic phenotype, metastasis, aggressiveness, resistance to chemotherapy, and immunotherapeutic approaches of SCLC. Different types of epigenetic modifications (DNA/histone methylation or acetylation) that can serve as predictive biomarkers for prognostication, treatment stratification, neuroendocrine lineage determination, and development of potential SCLC therapies are also discussed. We also review the utility of epigenetic targets/epidrugs in combination with first-line chemotherapy and immunotherapy that are currently under investigation in preclinical and clinical studies. Altogether, the information presents the inclusive landscape of SCLC epigenetics and epidrugs that will help translate the knowledge of epigenetics to improve SCLC outcomes.
- Published
- 2022
7. Histidine Enhances the Anticancer Effect of Gemcitabine against Pancreatic Cancer via Disruption of Amino Acid Homeostasis and Oxidant—Antioxidant Balance
- Author
-
Narendra Kumar, Satyanarayana Rachagani, Gopalakrishnan Natarajan, Alexandra Crook, Thiyagarajan Gopal, Vinothkumar Rajamanickam, Jyoti B. Kaushal, Sirpu N. Nagabhishek, Robert Powers, Surinder K. Batra, and Viswanathan Saraswathi
- Subjects
Cancer Research ,Oncology ,pancreatic cancer ,histidine ,gemcitabine ,glutathione ,hydrogen peroxide ,metabolomics - Abstract
Due to the severe toxicity posed by chemotherapeutic drugs, adjuvant nutritional intervention has gained increased attention in the treatment of pancreatic cancer (PC). Amino acid (AA) metabolism is aberrantly regulated in PC and circulating histidine (His) levels are low in PC patients. We hypothesized that His uptake and/or metabolism is dysregulated in PC and that combining His with gemcitabine (Gem), a drug used in the treatment of PC, will enhance the anti-cancer effects of Gem. We performed in vitro and in vivo studies to determine the anticancer effect of the combination of His and Gem against lethal PC. We demonstrate that circulating His levels are low in both human subjects and genetically engineered mice exhibiting pancreatic tumors. Interestingly, the expression of histidine ammonia lyase, an enzyme involved in His catabolism, is higher in PC compared to normal subjects. His + Gem exerts a more potent cytotoxic effect in PC cells compared to individual treatments. His treatment results in a profound increase in His accumulation, accompanied by a depletion of a number of AAs, promoting cancer cell survival and/or glutathione (GSH) synthesis. His but not Gem increases hydrogen peroxide and depletes cellular GSH. Supplementation with GSH protects cells against His + Gem-induced cytotoxicity. Further, our in vivo studies demonstrate that His + Gem potently reduced tumor mass and improved mouse survival. Taken together, our data suggest that PC cells exhibit an aberrant His uptake/accumulation which, in turn, leads to oxidative stress and depletion of AA pool, thereby enhancing the anticancer effect of Gem.
- Published
- 2023
- Full Text
- View/download PDF
8. MUC16 Promotes Liver Metastasis of Pancreatic Ductal Adenocarcinoma by Upregulating NRP2-Associated Cell Adhesion
- Author
-
Saravanakumar Marimuthu, Imayavaramban Lakshmanan, Sakthivel Muniyan, Shailendra K. Gautam, Rama Krishna Nimmakayala, Sanchita Rauth, Pranita Atri, Ashu Shah, Namita Bhyravbhatla, Kavita Mallya, Paul M. Grandgenett, Michael A. Hollingsworth, Kaustubh Datta, Maneesh Jain, Moorthy P. Ponnusamy, and Surinder K. Batra
- Subjects
Cancer Research ,Liver Neoplasms ,Membrane Proteins ,Adenocarcinoma ,Article ,Neuropilin-2 ,Pancreatic Neoplasms ,Mice ,Oncology ,Cell Movement ,CA-125 Antigen ,Cell Line, Tumor ,Cell Adhesion ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Carcinoma, Pancreatic Ductal ,Cell Proliferation - 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.
- Published
- 2022
9. MicroRNA-1: Diverse role of a small player in multiple cancers
- Author
-
Nivetha Sarah Ebenezer, Jawed A. Siddiqui, Mohd W. Nasser, Imayavaramban Lakshmanan, Ravi Salgia, Surinder K. Batra, Shailendra Kumar Maurya, and Parvez Khan
- Subjects
Male ,0301 basic medicine ,Tumor suppressor gene ,Angiogenesis ,Apoptosis ,Biology ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,microRNA ,medicine ,Humans ,Gene ,Cell Proliferation ,Neovascularization, Pathologic ,Mechanism (biology) ,High-Throughput Nucleotide Sequencing ,Cancer ,Cell Biology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,The Hallmarks of Cancer ,Cancer research ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The process of cancer initiation and development is a dynamic and complex mechanism involving multiple genetic and non-genetic variations. With the development of high throughput techniques like next-generation sequencing, the field of cancer biology extended beyond the protein-coding genes. It brought the functional role of noncoding RNAs into cancer-associated pathways. MicroRNAs (miRNAs) are one such class of noncoding RNAs regulating different cancer development aspects, including progression and metastasis. MicroRNA-1 (miR-1) is a highly conserved miRNA with a functional role in developing skeletal muscle precursor cells and cardiomyocytes, and acts as a consistent tumor suppressor gene. In humans, two discrete genes, MIR1–1 located on 20q13.333 and MIR1–2 located on 18q11.2 loci, encode for a single mature miR-1. Dysregulation or downregulation of miR-1 has been demonstrated in multiple cancers, including lung, breast, liver, prostate, colorectal, pancreatic, medulloblastoma, and gastric cancer. A vast number of studies have shown that miR-1 affects the multiple hallmarks of cancer like proliferation, invasion and metastasis, apoptosis, angiogenesis, chemosensitization, and immune modulation. The potential therapeutic applications of miR-1 in multiple cancer pathways provide a novel platform for developing anticancer therapies. This review focuses on the different antitumorigenic and therapeutic aspects of miR-1 including how it regulates tumor development and associated immunogenic functions.
- Published
- 2022
10. Pathophysiological role of growth differentiation factor 15 (GDF15) in obesity, cancer, and cachexia
- Author
-
Mohd W. Nasser, Parvez Khan, Surinder K. Batra, Maneesh Jain, Gunjan Sharma, Jawed A. Siddiqui, Ramesh Pothuraju, Parthasarathy Seshacharyulu, and Sakthivel Muniyan
- Subjects
Cachexia ,Glial Cell Line-Derived Neurotrophic Factor Receptors ,Growth Differentiation Factor 15 ,Endocrinology, Diabetes and Metabolism ,Immunology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Neurotrophic factors ,Neoplasms ,Tumor Microenvironment ,medicine ,Glial cell line-derived neurotrophic factor ,Humans ,Immunology and Allergy ,Obesity ,Tumor microenvironment ,biology ,business.industry ,Cancer ,medicine.disease ,biology.protein ,Cancer research ,GDF15 ,business ,Transforming growth factor - Abstract
Growth differentiation factor 15 or macrophage inhibitory cytokine-1 (GDF15/MIC-1) is a divergent member of the transforming growth factor β superfamily and has a diverse pathophysiological role in cancers, cardiometabolic disorder, and other diseases. GDF15 can control hematopoietic growth, energy homeostasis, adipose tissue metabolism, body growth, bone remodeling, and response to stress signals. The role of GDF15 in cancer development and progression is complicated and depends on the specific cancer type, stage, and tumor microenvironment. Recently, research on GDF15 and GDF15-associated signaling has accelerated due to the identification of the GDF15 receptor: glial cell line-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL). Therapeutic interventions to target GDF15 and/or GFRAL revealed the mechanisms that drive its activity and might improve overall outcomes of patients with metabolic disorders or cancer. The present review highlights the structure and functions of GDF15 and its receptor, emphasizing the pleiotropic role of GDF15 in obesity, tumorigenesis, metastasis, immunomodulation, and cachexia.
- Published
- 2022
11. The GSK3 kinase and LZTR1 protein regulate the stability of Ras family proteins and the proliferation of pancreatic cancer cells
- Author
-
Chitra Palanivel, Neha Chaudhary, Parthasarathy Seshacharyulu, Jesse L. Cox, Ying Yan, Surinder K. Batra, and Michel M. Ouellette
- Subjects
Cancer Research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Pancreatic cancer ,GSK3 ,Pancreatic Neoplasms ,Glycogen Synthase Kinase 3 ,Protein stability ,ras Proteins ,Humans ,LZTR1 ,RC254-282 ,Cell Proliferation ,Signal Transduction ,Transcription Factors ,RAS - Abstract
Ras family proteins are membrane-bound GTPases that control proliferation, survival, and motility. Many forms of cancers are driven by the acquisition of somatic mutations in a RAS gene. In pancreatic cancer (PC), more than 90% of tumors carry an activating mutation in KRAS. Mutations in components of the Ras signaling pathway can also be the cause of RASopathies, a group of developmental disorders. In a subset of RASopathies, the causal mutations are in the LZTR1 protein, a substrate adaptor for E3 ubiquitin ligases that promote the degradation of Ras proteins. Here, we show that the function of LZTR1 is regulated by the glycogen synthase kinase 3 (GSK3). In PC cells, inhibiting or silencing GSK3 led to a decline in the level of Ras proteins, including both wild type Ras proteins and the oncogenic Kras protein. This decline was accompanied by a 3-fold decrease in the half-life of Ras proteins and was blocked by the inhibition of the proteasome or the knockdown of LZTR1. Irrespective of the mutational status of KRAS, the decline in Ras proteins was observed and accompanied by a loss of cell proliferation. This loss of proliferation was blocked by the knockdown of LZTR1 and could be recapitulated by the silencing of either KRAS or GSK3. These results reveal a novel GSK3-regulated LZTR1-dependent mechanism that controls the stability of Ras proteins and proliferation of PC cells. The significance of this novel pathway to Ras signaling and its contribution to the therapeutic properties of GSK3 inhibitors are both discussed.
- Published
- 2022
12. MUC16 and TP53 family co-regulate tumor-stromal heterogeneity in pancreatic adenocarcinoma
- Author
-
Ramakanth Chirravuri-Venkata, Vi Dam, Rama Krishna Nimmakayala, Zahraa Wajih Alsafwani, Namita Bhyravbhatla, Imayavaramban Lakshmanan, Moorthy P. Ponnusamy, Sushil Kumar, Maneesh Jain, Dario Ghersi, and Surinder K. Batra
- Subjects
Cancer Research ,Oncology - Abstract
MUC16/CA125 is one of the few oldest cancer biomarkers still used in current clinical practice. As mesothelium is an abundant source of MUC16 and a major contributor to stromal heterogeneity in PDAC, we investigated the regulation of MUC16 in tumor and stromal compartments individually. The trajectories constructed using the single-cell transcriptomes of stromal cells from KPC tumors demonstrated continuity in the trajectory path between MUC16-expressing mesothelial cells and other CAF subsets. Further, the tumor tissues of MUC16 whole-body knockout (KPCM) showed dysregulation in the markers of actomyosin assembly and fibroblast differentiation (iCAF and myCAF), indicating that MUC16 has an extra-tumoral role in controlling CAF differentiation. Although we found mesothelium-derivative stromal cells to be bystanders in normal pancreas, the proportion of these cells was higher in invasive PDAC, particularly in TP53 deficient tumors. Moreover, we also detail the regulation of MUC16, KRAS, and SOX9 by TP53 family members (TP53 and TP63) using multi-omics data from knockout models, PDAC cell lines, and human PDAC tissues.
- Published
- 2023
13. The Mucin Family of Proteins: Candidates as Potential Biomarkers for Colon Cancer
- Author
-
Kristin E. Cox, Shanglei Liu, Thinzar M. Lwin, Robert M. Hoffman, Surinder K. Batra, and Michael Bouvet
- Subjects
Cancer Research ,adenocarcinoma ,Prevention ,Oncology and Carcinogenesis ,colorectal cancer ,hyperplastic polyps ,mucins ,serrated polyps ,Colo-Rectal Cancer ,Rare Diseases ,Oncology ,prognostics ,mucinous carcinoma ,adenoma ,adenomatous polyps ,Digestive Diseases ,Cancer - Abstract
Mucins (MUC1–MUC24) are a family of glycoproteins involved in cell signaling and barrier protection. They have been implicated in the progression of numerous malignancies including gastric, pancreatic, ovarian, breast, and lung cancer. Mucins have also been extensively studied with respect to colorectal cancer. They have been found to have diverse expression profiles amongst the normal colon, benign hyperplastic polyps, pre-malignant polyps, and colon cancers. Those expressed in the normal colon include MUC2, MUC3, MUC4, MUC11, MUC12, MUC13, MUC15 (at low levels), and MUC21. Whereas MUC5, MUC6, MUC16, and MUC20 are absent from the normal colon and are expressed in colorectal cancers. MUC1, MUC2, MUC4, MUC5AC, and MUC6 are currently the most widely covered in the literature regarding their role in the progression from normal colonic tissue to cancer.
- Published
- 2023
14. Tumor microenvironment enriches the stemness features: the architectural event of therapy resistance and metastasis
- Author
-
Palanisamy, Nallasamy, Rama Krishna, Nimmakayala, Seema, Parte, Abhirup C, Are, Surinder K, Batra, and Moorthy P, Ponnusamy
- Subjects
Cancer Research ,Oncology ,Molecular Medicine - Abstract
Cancer divergence has many facets other than being considered a genetic term. It is a tremendous challenge to understand the metastasis and therapy response in cancer biology; however, it postulates the opportunity to explore the possible mechanism in the surrounding tumor environment. Most deadly solid malignancies are distinctly characterized by their tumor microenvironment (TME). TME consists of stromal components such as immune, inflammatory, endothelial, adipocytes, and fibroblast cells. Cancer stem cells (CSCs) or cancer stem-like cells are a small sub-set of the population within cancer cells believed to be a responsible player in the self-renewal, metastasis, and therapy response of cancer cells. The correlation between TME and CSCs remains an enigma in understanding the events of metastasis and therapy resistance in cancer biology. Recent evidence suggests that TME dictates the CSCs maintenance to arbitrate cancer progression and metastasis. The immune, inflammatory, endothelial, adipocyte, and fibroblast cells in the TME release growth factors, cytokines, chemokines, microRNAs, and exosomes that provide cues for the gain and maintenance of CSC features. These intricate cross-talks are fueled to evolve into aggressive, invasive, migratory phenotypes for cancer development. In this review, we have abridged the recent developments in the role of the TME factors in CSC maintenance and how these events influence the transition of tumor progression to further translate into metastasis and therapy resistance in cancer.
- Published
- 2022
15. The tumor microenvironment as driver of stemness and therapeutic resistance in breast cancer: New challenges and therapeutic opportunities
- Author
-
Mohd W. Nasser, Abid Hamid, Nissar A. Wani, Umar Mehraj, Raid Al-Baradie, Manzoor Ahmad Mir, Mohammad Haris, Ajaz A. Bhat, Bader Alshehri, Mohammed A. Zargar, Muzafar A. Macha, Rais A. Ganai, and Surinder K. Batra
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,business.industry ,medicine.medical_treatment ,Cancer ,General Medicine ,Immunotherapy ,medicine.disease ,Metastasis ,Radiation therapy ,Breast cancer ,Oncology ,Cancer cell ,medicine ,Cancer research ,Molecular Medicine ,sense organs ,business - Abstract
Breast cancer (BC), the second most common cause of cancer-related deaths, remains a significant threat to the health and wellness of women worldwide. The tumor microenvironment (TME), comprising cellular components, such as cancer-associated fibroblasts (CAFs), immune cells, endothelial cells and adipocytes, and noncellular components such as extracellular matrix (ECM), has been recognized as a critical contributor to the development and progression of BC. The interplay between TME components and cancer cells promotes phenotypic heterogeneity, cell plasticity and cancer cell stemness that impart tumor dormancy, enhanced invasion and metastasis, and the development of therapeutic resistance. While most previous studies have focused on targeting cancer cells with a dismal prognosis, novel therapies targeting stromal components are currently being evaluated in preclinical and clinical studies, and are already showing improved efficacies. As such, they may offer better means to eliminate the disease effectively. In this review, we focus on the evolving concept of the TME as a key player regulating tumor growth, metastasis, stemness, and the development of therapeutic resistance. Despite significant advances over the last decade, several clinical trials focusing on the TME have failed to demonstrate promising effectiveness in cancer patients. To expedite clinical efficacy of TME-directed therapies, a deeper understanding of the TME is of utmost importance. Secondly, the efficacy of TME-directed therapies when used alone or in combination with chemo- or radiotherapy, and the tumor stage needs to be studied. Likewise, identifying molecular signatures and biomarkers indicating the type of TME will help in determining precise TME-directed therapies.
- Published
- 2021
16. Small molecule antagonist of CXCR2 and CXCR1 inhibits tumor growth, angiogenesis, and metastasis in pancreatic cancer
- Author
-
Dipakkumar R. Prajapati, Caitlin Molczyk, Abhilasha Purohit, Sugandha Saxena, Reegan Sturgeon, Bhavana J. Dave, Sushil Kumar, Surinder K. Batra, and Rakesh K. Singh
- Subjects
Cancer Research ,Oncology - Published
- 2023
17. MASTL regulates EGFR signaling to impact pancreatic cancer progression
- Author
-
Punita Dhawan, Moorthy P. Ponnusamy, Surinder K. Batra, Shailender S. Chauhan, Lynette M. Smith, Jaya Prakash Uppada, Geoffrey A. Talmon, Susmita Barman, Satyanarayana Rachagani, Amar B. Singh, Iram Fatima, and Sanchita Rauth
- Subjects
Cancer Research ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Malignancy ,Deoxycytidine ,Article ,Serine ,Mice ,Loss of Function Mutation ,Cell Line, Tumor ,Pancreatic cancer ,Genetics ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Molecular Biology ,biology ,Protein Stability ,Kinase ,Cancer ,medicine.disease ,Gemcitabine ,Up-Regulation ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Gain of Function Mutation ,Disease Progression ,Cancer research ,biology.protein ,KRAS ,Microtubule-Associated Proteins ,Neoplasm Transplantation ,Signal Transduction ,medicine.drug - Abstract
Pancreatic cancer (PC) remains a major cause of cancer-related deaths primarily due to its inherent potential of therapy resistance. Checkpoint inhibitors have emerged as promising anti-cancer agents when used in combination with conventional anti-cancer therapies. Recent studies have highlighted a critical role of the Greatwall kinase (MASTL; Microtubule-associated serine/threonine-protein kinase-like) in promoting oncogenic malignancy and resistance to anti-cancer therapies; however, its role in PC remains unknown. Based on a comprehensive investigation involving PC patient samples, murine models of PC progression (Kras;PdxCre-KC and Kras;p53;PdxCre-KPC), and loss and gain of function studies, we report a previously undescribed critical role of MASTL in promoting cancer malignancy and therapy resistance. Mechanistically, MASTL promotes PC by modulating the epidermal growth factor receptor (EGFR) protein stability and, thereupon, kinase signaling. We further demonstrate that combinatorial therapy targeting MASTL promotes the efficacy of the cell-killing effects of Gemcitabine using both genetic and pharmacological inhibitions. Taken together, this study identifies a key role of MASTL in promoting PC progression and its utility as a novel target in promoting sensitivity to the anti-pancreatic cancer therapies.
- Published
- 2021
18. Protein Phosphatase 2A as a Therapeutic Target in Small Cell Lung Cancer
- Author
-
Tamara, Mirzapoiazova, Gang, Xiao, Bolot, Mambetsariev, Mohd W, Nasser, Emily, Miaou, Sharad S, Singhal, Saumya, Srivastava, Isa, Mambetsariev, Michael S, Nelson, Arin, Nam, Amita, Behal, Leonidas, Arvanitis, Pranita, Atri, Markus, Muschen, François L H, Tissot, James, Miser, John S, Kovach, Martin, Sattler, Surinder K, Batra, Prakash, Kulkarni, and Ravi, Salgia
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Phosphatase ,Antineoplastic Agents ,Apoptosis ,Gene Expression Regulation, Enzymologic ,Piperazines ,Article ,chemistry.chemical_compound ,Atezolizumab ,Tumor Cells, Cultured ,medicine ,Humans ,Protein Phosphatase 2 ,Viability assay ,Cell Proliferation ,Protein phosphatase 2 ,Immunotherapy ,Small Cell Lung Carcinoma ,Carboplatin ,Gene Expression Regulation, Neoplastic ,Endothelial stem cell ,Oncology ,chemistry ,Cancer research - Abstract
Protein phosphatase 2A (PP2A), a serine/threonine phosphatase involved in the regulation of apoptosis, proliferation, and DNA-damage response, is overexpressed in many cancers, including small cell lung cancer (SCLC). Here we report that LB100, a small molecule inhibitor of PP2A, when combined with platinum-based chemotherapy, synergistically elicited an antitumor response both in vitro and in vivo with no apparent toxicity. Using inductively coupled plasma mass spectrometry, we determined quantitatively that sensitization via LB100 was mediated by increased uptake of carboplatin in SCLC cells. Treatment with LB100 alone or in combination resulted in inhibition of cell viability in two-dimensional culture and three-dimensional spheroid models of SCLC, reduced glucose uptake, and attenuated mitochondrial and glycolytic ATP production. Combining LB100 with atezolizumab increased the capacity of T cells to infiltrate and kill tumor spheroids, and combining LB100 with carboplatin caused hyperphosphorylation of the DNA repair marker γH2AX and enhanced apoptosis while attenuating MET signaling and invasion through an endothelial cell monolayer. Taken together, these data highlight the translational potential of inhibiting PP2A with LB100 in combination with platinum-based chemotherapy and immunotherapy in SCLC.
- Published
- 2021
19. Neuropilin-2 regulates androgen-receptor transcriptional activity in advanced prostate cancer
- Author
-
Samikshan Dutta, Navatha Shree Polavaram, Ridwan Islam, Sreyashi Bhattacharya, Sanika Bodas, Thomas Mayr, Sohini Roy, Sophie Alvarez Y. Albala, Marieta I. Toma, Anza Darehshouri, Angelika Borkowetz, Stefanie Conrad, Susanne Fuessel, Manfred Wirth, Gustavo B. Baretton, Lorenz C. Hofbauer, Paramita Ghosh, Kenneth J. Pienta, David L. Klinkebiel, Surinder K. Batra, Michael H. Muders, and Kaustubh Datta
- Subjects
Male ,Urologic Diseases ,Cancer Research ,Aging ,Clinical Sciences ,Oncology and Carcinogenesis ,Castration-Resistant ,Article ,Cell Line ,Androgen ,Cell Line, Tumor ,Receptors ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Oncology & Carcinogenesis ,Aetiology ,Molecular Biology ,Cancer ,Tumor ,Prostate Cancer ,Prostatic Neoplasms ,Neuropilin-2 ,Prostatic Neoplasms, Castration-Resistant ,Receptors, Androgen ,5.1 Pharmaceuticals ,Androgens ,Development of treatments and therapeutic interventions ,Signal Transduction - Abstract
Aberrant transcriptional activity of androgen receptor (AR) is one of the dominant mechanisms for developing of castration-resistant prostate cancer (CRPC). Analyzing AR-transcriptional complex related to CRPC is therefore important towards understanding the mechanism of therapy-resistance. While studying its mechanism, we observed that a transmembrane protein called neuropilin-2 (NRP2) plays a contributory role in forming a novel AR-transcriptional complex containing nuclear pore proteins. Using immunogold electron microscopy, high-resolution confocal microscopy, chromatin immunoprecipitation, proteomics, and other biochemical techniques, we delineated the molecular mechanism of how a specific splice variant of NRP2 becomes sumoylated upon ligand stimulation and translocates to the inner nuclear membrane. This splice variant of NRP2 then stabilizes the complex between AR and nuclear pore proteins to promote CRPC specific gene expression. Both full-length and splice variants of AR have been identified in this specific transcriptional complex. In vitro cell line-based assays indicated that depletion of NRP2 not only destabilizes the AR-nuclear pore protein interaction but also inhibits the transcriptional activities of AR. Using an in vivo bone metastasis model, we showed that the inhibition of NRP2 led to the sensitization of CRPC cells toward established anti-AR therapies such as enzalutamide. Overall, our finding emphasize the importance of combinatorial inhibition of NRP2 and AR as an effective therapeutic strategy against treatment refractory prostate cancer.
- Published
- 2022
20. Small molecule inhibitor against onco-mucins disrupts Src/FosL1 axis to enhance gemcitabine efficacy in pancreatic ductal adenocarcinoma
- Author
-
Chunmeng Zhang, Pranita Atri, Palanisamy Nallasamy, Seema Parte, Sanchita Rauth, Rama Krishna Nimmakayala, Saravanakumar Marimuthu, Ramakanth Chirravuri-Venkata, Rakesh Bhatia, Sushanta Halder, Ashu Shah, Jesse L. Cox, Lynette Smith, Sushil Kumar, Jason M. Foster, Rakesh C. Kukreja, Parthasarathy Seshacharyulu, Moorthy P. Ponnusamy, and Surinder K. Batra
- Subjects
Cancer Research ,Oncology - Abstract
Mucin MUC4 is an aberrantly expressed oncogene in pancreatic ductal adenocarcinoma (PDAC), yet no pharmacological inhibitors have been identified to target MUC4. Here, we adapted an in silico screening method using the Cancer Therapeutic Response Database (CTRD) to Identify Small Molecule Inhibitors against Mucins (SMIMs). We identified Bosutinib as a candidate drug to target oncogenic mucins among 126 FDA-approved drugs from CTRD screening. Functionally, Bosutinib treatment alone/and in combination with gemcitabine (Gem)/5' fluorouracil (5FU) reduced in vitro viability, migration, and colony formation in multiple PDAC cell lines as well as human PDAC organoid prolifertaion and growth and in vivo xenograft growth. Further, biochemical and molecular analyses showed that Bosutinib exhibited these functional effects by downregulating MUC4 mucin at both transcript and translation levels in a dose- and time-dependent manner. Mechanistically, global transcriptome analysis in PDAC cells upon treatment with Bosutinib revealed disruption of the Src-ERK/AKT-FosL1 pathway, leading to decreased expression of MUC4 and MUC5AC mucins. Taken together, Bosutinib is a promising, novel, and highly potent SMIMs to target MUC4/MUC5AC mucins. This mucin-targeting effect of Bosutinib can be exploited in the future with cytotoxic agents to treat mucinous tumors.
- Published
- 2022
21. ST6GalNAc‐I promotes lung cancer metastasis by altering MUC5AC sialylation
- Author
-
Moorthy P. Ponnusamy, Rohitesh Gupta, Subodh M. Lele, Sanchita Rauth, Saravanakumar Marimuthu, Mohd W. Nasser, Rahat Jahan, Imayavaramban Lakshmanan, Kavita Mallya, Raghupathy Vengoji, Sanjib Chaudhary, Pranita Atri, Surinder K. Batra, Ravi Salgia, Parthasarathy Seshacharyulu, Apar Kishor Ganti, Joseph Carmicheal, Lynette M. Smith, Ramakanth Chirravuri-Venkata, Naveenkumar Perumal, Sukhwinder Kaur, and Satyanarayana Rachagani
- Subjects
0301 basic medicine ,Cancer Research ,Glycosylation ,Lung Neoplasms ,integrin β4 ,Mucin 5AC ,Metastasis ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Humans ,RC254-282 ,Research Articles ,chemistry.chemical_classification ,FAK ,Mucin ,Liver Neoplasms ,Colocalization ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell migration ,General Medicine ,medicine.disease ,MUC5AC ,ST6GalNAc‐I ,N-Acetylneuraminic Acid ,Sialyltransferases ,Sialic acid ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,lung cancer metastasis ,Molecular Medicine ,Ectopic expression ,Glycoprotein ,Research Article - 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 KrasG12D/+; 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., In the presence of mutant p53R175H, ST6GalNAc‐I mediates MUC5AC sialylation that increases the aggressiveness of lung adenocarcinoma (LUAD). MUC5AC sialylation is required for integrin β4 interaction that induces angiogenesis and liver metastasis. Hence, targeting the ST6GalNAc‐I/MUC5AC could prevent LUAD metastasis.
- Published
- 2021
22. Mucins reprogram stemness, metabolism and promote chemoresistance during cancer progression
- Author
-
Saravanakumar Marimuthu, Imayavaramban Lakshmanan, Chunmeng Zhang, Koelina Ganguly, Sanchita Rauth, Moorthy P. Ponnusamy, and Surinder K. Batra
- Subjects
0301 basic medicine ,Cancer Research ,Population ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Neoplasms ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Neoplasm Metastasis ,education ,education.field_of_study ,Mucin ,Mucins ,Cancer ,Cellular Reprogramming ,medicine.disease ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Neoplastic Stem Cells ,Cancer research ,Carcinogenesis ,Signal Transduction - Abstract
BACKGROUND: Mucins are high molecular weight glycoproteins dysregulated in aggressive cancers. The role of mucins in disease progression, tumor proliferation, and chemotherapy resistance has been studied extensively. MAIN BODY: This article provides a comprehensive review of mucin’s function as a physical barrier and the implication of mucin overexpression in impeded drug delivery to solid tumors. Mucins regulate the epithelial to mesenchymal transition (EMT) of cancer cells via several canonical and non-canonical oncogenic signaling pathways. Furthermore, mucins play an extensive role in enriching and maintaining the cancer stem cell (CSC) population, thereby sustaining the self-renewing and chemoresistant cellular pool in the bulk tumor. It has recently been demonstrated that mucins regulate the metabolic reprogramming during oncogenesis and cancer progression, which account for tumor cell survival, proliferation, and drug-resistance. CONCLUSION: This review article focuses on delineating mucin’s role in oncogenic signaling and aberrant regulation of gene expressions, culminating in CSC maintenance, metabolic rewiring, and development of chemoresistance, tumor progression, and metastasis.
- Published
- 2021
23. Mucins as contrast agent targets for fluorescence-guided surgery of pancreatic cancer
- Author
-
Kathryn M. Muilenburg, Carly C. Isder, Prakash Radhakrishnan, Surinder K. Batra, Quan P. Ly, Mark A. Carlson, Michael Bouvet, Michael A. Hollingsworth, and Aaron M. Mohs
- Subjects
Cancer Research ,Oncology - Published
- 2023
24. Epigenetic regulation of cancer-associated fibroblast heterogeneity
- Author
-
Rachel J. Kehrberg, Namita Bhyravbhatla, Surinder K. Batra, and Sushil Kumar
- Subjects
Cancer Research ,Oncology ,Genetics - Published
- 2023
25. Abstract 1756: Targeting LIFR/c-Myc Axis to Overcome Docetaxel Resistance in Prostate Cancer
- Author
-
Sushanta Halder, Sakthivel Muniyan, Ramakanth Chirravuri-Venkata, Rama Krishna Nimmakayala, Palanisamy Nallasamy, Hareesh B. Nair, Moorthy P Ponnusamy, Surinder K. Batra, and Parthasarathy Seshacharyulu
- Subjects
Cancer Research ,Oncology - Abstract
Background: Docetaxel has been the most effective chemotherapeutic option after the emergence of Castration-resistant prostate cancer (CRPC). However, more than 50% of patients develop Docetaxel Resistance (DoceR) within three years of treatment. It also shows severe adverse effects that lead to dose reduction and treatment failure. Thus, targeting the underlying mechanism of DoceR could improve survival benefits for CRPC patients. In this study, we identified leukemia inhibitory factor receptor (LIFR) as a candidate for DoceR. We targeted it with EC914, a first-in-class oral small molecule, to overcome DoceR in CRPC. Methods: We developed three DoceR (PC3-R60, 22Rv1-R110, and LNCaP(C-83)-R50) isogeneic sub-lines by chronic Docetaxel treatment for six months. We analyzed those cell lines for drug resistance markers using RT-PCR and western blot. We evaluated the effect of EC914 in overcoming DoceR using cytotoxicity assay in Incucyte® live imaging system, colony survival assay, apoptosis induction, and cell cycle arrest using FACS analysis. RNA-seq analysis revealed molecular pathways related to EC914-mediated Docetaxel- sensitivity. In addition, we utilized RT-PCR and western blot analysis to examine EC914-responsive cancer stem cell (CSC) and LIFR/STAT pathway-related genes and proteins. Finally, we adopted human and mouse syngeneic PCa cells and patient-derived xenograft models to test the in vivo efficacy of EC914 in combination with Docetaxel. Results: LIFR showed significantly high mRNA and protein expression in Docetaxel resistant PCa cell model. EC914 combined with Docetaxel reduced considerably in vitro tumorigenicity (proliferation (p Conclusion: For the first time, we identified c-Myc pathway genes as potential LIFR signaling targets to overcome docetaxel resistance. Our findings support a new role and mechanism(s) of LIFR in CRPC and could inhibit MYC-addicted tumors. Citation Format: Sushanta Halder, Sakthivel Muniyan, Ramakanth Chirravuri-Venkata, Rama Krishna Nimmakayala, Palanisamy Nallasamy, Hareesh B. Nair, Moorthy P Ponnusamy, Surinder K. Batra, Parthasarathy Seshacharyulu. Targeting LIFR/c-Myc Axis to Overcome Docetaxel Resistance in Prostate Cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1756.
- Published
- 2023
26. Abstract 5850: Development and characterization of patient-derived pancreatic fibroblasts to study CAF heterogeneity
- Author
-
Nidhi V. Dwivedi, Shailendra K. Gautam, Satya Rachagani, Ramakanth Venkata, Vipin Dalal, Maneesh Jain, and Surinder K. Batra
- Subjects
Cancer Research ,Oncology - Abstract
Pancreatic ductal adenocarcinoma (PDAC) has complex tumor microenvironment (TME), characterized by phenotypically and functionally heterogenous cancer-associated fibroblasts (CAFs). Different CAF subtypes have been identified that evidently contribute to desmoplasia, immunosuppression, and therapy response variably. The lack of models to study their plasticity and heterogeneity reproducibly poses a major limitation. We, thus, describe the development and characterization of a panel of immortalized patient-derived pancreatic fibroblasts (iPDPFs), studying their impact on PDAC cell lines in vitro and in vivo. Method: We developed six iPDPF lines using human telomerase reverse transcriptase (hTERT) from cancer-adjacent normal pancreas (9-26-NP), chronic pancreatitis (CPP1), and PDAC CAFs (9-17-P, 10-15-P, 10-03-P, & 10-32-P). Immunoblotting, bulk-RNAseq, and qPCR were performed for their molecular characterization. Functional characterization was done in vitro using collagen contraction assay and co-culture studies with COLO357. The impact of iPDPFs on the tumor cell behavior in vivo was studied by their co-implantation with COLO357 in athymic nude mice. Results: The iPDPFs exhibited variable expression of activation, inflammatory, and ECM markers indicative of heterogeneity across the panel. While CAFs expressed high levels of αSMA (alpha-smooth muscle actin) and fibroblast-specific protein-1 (FSP1), the expression of these markers was low in 9-26 NP & CPP-1. Also, the iPDPFs variably expressed TGFβ, PDGFRβ, VDR, COL1A, and small GTPases. High expression of inflammatory markers was observed in 9-26 NP, CPP-1, & 10-32-P suggesting their iCAF type while 9-17-P, 10-03-P, and 10-15-P were of myCAF type. These observations were recapitulated in the transcriptomic analysis. Interestingly, the marker expression changed with passage suggesting existence of plasticity in the iPDPFs. 9-17-P induced highest collagen contraction (47.9%) while 10-15-P induced the least contraction (25.89%). Co-culture of iPDPFs with COLO357 enhanced the expression of ROCK-1. In vivo, iPDPFs resulted in stroma-rich tumors; however, the extent of stroma and growth rate were variable across iPDPF lines and implantation sites. 9-17-P xenograft tumors exhibited the highest percent αSMA (14.5%), and FSP1 (30.7%) positive cells. Limited analysis showed that iPDPF co-implanted tumors exhibited variable vascularity and ECM composition suggesting that the heterogeneity observed in vitro was manifested in vivo. Conclusion: Overall, the iPDPFs recapitulate pancreatic fibroblasts heterogeneity, reflecting the overlapping nature of CAF subtypes. Also, the iPDPFs are functionally variable and plastic in vitro. Importantly, these fibroblasts exert variable effects on the tumor cells in vivo. The iPDPFs can thus serve as a useful model to reproducibly study CAF heterogeneity and plasticity in PDAC. Citation Format: Nidhi V. Dwivedi, Shailendra K. Gautam, Satya Rachagani, Ramakanth Venkata, Vipin Dalal, Maneesh Jain, Surinder K. Batra. Development and characterization of patient-derived pancreatic fibroblasts to study CAF heterogeneity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5850.
- Published
- 2023
27. Abstract 1283: MUC5AC promotes brain metastases and colonization in non small cell lung cancer
- Author
-
Sanjib Chaudhary, Shailendra Kumar Maurya, Jawed Siddiqui, Mohd W. Nasser, Surinder K. Batra, Imayavaramban Lakshmanan, and Apar Kishor Ganti
- Subjects
Cancer Research ,Oncology - Abstract
Background: Non-small cell lung adenocarcinoma (NSCLC), constitutes 80-85% of the lung cancers. The 5-year survival of patients with metastatic lung cancer is only 5%. Additionally, the prognosis worsens in NSCLC patients (40-50%) that have developed brain metastasis. Mucin 5AC (MUC5AC) is overexpressed in the primary NSCLC and brain metastatic tissues; however, the mechanism in the MUC5AC-mediated brain metastasis is unclear. Methods: Immunohistochemistry was performed on lung cancer samples. Expression studies were conducted in the brain-tropic cell lines (A549-BrM, PC9-BrM, and HCC4006-BrM cells) and their respective parental lines. CRISPR Cas9 or shRNA was used to either knockout/knockdown MUC5AC in the cell lines. Mass-spectrometry analysis and protein arrays were conducted in the brain trophic cells. In vivo mouse models involving intracardiac injection of cancer cells were performed, and the effect of MUC5AC brain metastasis and survival was analyzed. Results: MUC5AC was significantly increased in NSCLC brain metastatic tissues. Similarly, MUC5AC expression was elevated in NSCLC brain tropic (BrM) cell lines compared to parental lines. Treatment of BrM or parental cells with the conditioned media of normal human astrocytes resulted in increased MUC5AC levels. We observed astrocyte-derived CCL2-induced the MUC5AC expression in both parental and BrM cells. Mass-spectrometry based proteomic analysis indicated decreased cell adhesion molecules such as ALCAM, ITGB4, ITGB5, CDH17, and DSG2 in MUC5AC knockdown tropic cells. Further, Mass-spectrometry based interactome studies revealed MUC5AC interaction with Annexin A2 enabling the cancer cell adhesion. Intracardiac injection of MUC5AC knockdown A549-BrM (A549-BrM-shMUC5AC) cells showed a significant reduction in brain metastasis and increased survival of the mice compared to scramble control group (P Conclusion: Our study identified the role of MUC5AC in metastasis and colonization in the brain environment. Mechanistically, MUC5AC interacts with the Annexin A2 during metastasis, while astrocyte derived CCL2 further induced the MUC5AC expression facilitating the colonization of the NSCLC cells in the brain. Hence, targeting MUC5AC and its interactions partner Annexin A2 may prevent NSCLC brain metastasis. Citation Format: Sanjib Chaudhary, Shailendra Kumar Maurya, Jawed Siddiqui, Mohd W. Nasser, Surinder K. Batra, Imayavaramban Lakshmanan, Apar Kishor Ganti. MUC5AC promotes brain metastases and colonization in non small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1283.
- Published
- 2023
28. Mucins as Potential Biomarkers for Early Detection of Cancer
- Author
-
Shailendra K. Gautam, Parvez Khan, Gopalakrishnan Natarajan, Pranita Atri, Abhijit Aithal, Apar K. Ganti, Surinder K. Batra, Mohd W. Nasser, and Maneesh Jain
- Subjects
Cancer Research ,Oncology - Abstract
Early detection significantly correlates with improved survival in cancer patients. So far, a limited number of biomarkers have been validated to diagnose cancers at an early stage. Considering the leading cancer types that contribute to more than 50% of deaths in the USA, we discuss the ongoing endeavors toward early detection of lung, breast, ovarian, colon, prostate, liver, and pancreatic cancers to highlight the significance of mucin glycoproteins in cancer diagnosis. As mucin deregulation is one of the earliest events in most epithelial malignancies following oncogenic transformation, these high-molecular-weight glycoproteins are considered potential candidates for biomarker development. The diagnostic potential of mucins is mainly attributed to their deregulated expression, altered glycosylation, splicing, and ability to induce autoantibodies. Secretory and shed mucins are commonly detected in patients’ sera, body fluids, and tumor biopsies. For instance, CA125, also called MUC16, is one of the biomarkers implemented for the diagnosis of ovarian cancer and is currently being investigated for other malignancies. Similarly, MUC5AC, a secretory mucin, is a potential biomarker for pancreatic cancer. Moreover, anti-mucin autoantibodies and mucin-packaged exosomes have opened new avenues of biomarker development for early cancer diagnosis. In this review, we discuss the diagnostic potential of mucins in epithelial cancers and provide evidence and a rationale for developing a mucin-based biomarker panel for early cancer detection.
- Published
- 2023
29. Secretory Mucin 5AC Promotes Neoplastic Progression by Augmenting KLF4-Mediated Pancreatic Cancer Cell Stemness
- Author
-
Shiv Ram Krishn, Ramesh Pothuraju, Koelina Ganguly, Sudhua Ayala, Sukhwinder Kaur, Christopher M. Evans, Satyanarayana Rachagani, Sushil Kumar, Sanchita Rauth, Ashu Shah, Jesse L. Cox, Moorthy P. Ponnusamy, Rahat Jahan, Pranita Atri, Palanisamy Nallasamy, Surinder K. Batra, and Lynette M. Smith
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Population ,Kruppel-Like Transcription Factors ,Pancreatic Intraepithelial Neoplasia ,Apoptosis ,Mucin 5AC ,Biology ,medicine.disease_cause ,digestive system ,Article ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Pancreatic cancer ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,education ,Cell Proliferation ,Mice, Knockout ,education.field_of_study ,Mucin ,respiratory system ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,KLF4 ,030220 oncology & carcinogenesis ,Cancer cell ,Neoplastic Stem Cells ,Cancer research ,Female ,sense organs ,Carcinogenesis ,Carcinoma, Pancreatic Ductal - Abstract
Secreted mucin 5AC (MUC5AC) is the most abundantly overexpressed member of the mucin family during early pancreatic intraepithelial neoplasia stage I (PanIN-I) of pancreatic cancer. To comprehend the contribution of Muc5ac in pancreatic cancer pathology, we genetically ablated it in an autochthonous murine model (KrasG12D; Pdx-1cre, KC), which mirrors the early stages of pancreatic cancer development. Neoplastic onset and the PanIN lesion progression were significantly delayed in Muc5ac knockout (KrasG12D; Pdx-1 cre; Muc5ac−/−, KCM) animals with a 50% reduction in PanIN-2 and 70% reduction in PanIN-3 lesions compared with KC at 50 weeks of age. High-throughput RNA-sequencing analysis from pancreatic tissues of KCM animals revealed a significant decrease in cancer stem cell (CSC) markers Aldh1a1, Klf4, EpCAM, and CD133. Furthermore, the silencing of MUC5AC in human pancreatic cancer cells reduced their tumorigenic propensity, as indicated by a significant decline in tumor formation frequency by limiting dilution assay upon subcutaneous administration. The contribution of MUC5AC in CSC maintenance was corroborated by a significant decrease in tumor burden upon orthotopic implantation of MUC5AC-depleted pancreatic cancer cells. Mechanistically, MUC5AC potentiated oncogenic signaling through integrin αvβ5, pSrc (Y416), and pSTAT3 (Y705). Phosphorylated STAT3, in turn, upregulated Klf4 expression, thereby enriching the self-renewing CSC population. A strong positive correlation of Muc5ac with Klf4 and pSTAT3 in the PanIN lesions of KC mouse pancreas reinforces the crucial involvement of MUC5AC in bolstering the CSC-associated tumorigenic properties of Kras-induced metaplastic cells, which leads to pancreatic cancer onset and progression. Significance: This study elucidates that de novo expression of MUC5AC promotes cancer cell stemness during Kras-driven pancreatic tumorigenesis and can be targeted for development of a novel therapeutic regimen.
- Published
- 2021
30. The Current Landscape of Antibody-based Therapies in Solid Malignancies
- Author
-
Ashu Shah, Sukhwinder Kaur, Abhijit Aithal, Koelina Ganguly, Grish C. Varshney, Sanchita Rauth, Catherine Orzechowski, Surinder K. Batra, and Maneesh Jain
- Subjects
0301 basic medicine ,Tumor targeting ,medicine.drug_class ,Receptor expression ,Medicine (miscellaneous) ,challenges ,Review ,Monoclonal antibody ,Antibodies ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, Neoplasm ,Neoplasms ,medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,Animals ,Humans ,cancer ,Effector functions ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Tumor microenvironment ,therapy ,biology ,business.industry ,Receptors, IgG ,Antibodies, Monoclonal ,Tumor antigen ,mechanisms of action ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,business - Abstract
Over the past three decades, monoclonal antibodies (mAbs) have revolutionized the landscape of cancer therapy. Still, this benefit remains restricted to a small proportion of patients due to moderate response rates and resistance emergence. The field has started to embrace better mAb-based formats with advancements in molecular and protein engineering technologies. The development of a therapeutic mAb with long-lasting clinical impact demands a prodigious understanding of target antigen, effective mechanism of action, gene engineering technologies, complex interplay between tumor and host immune system, and biomarkers for prediction of clinical response. This review discusses the various approaches used by mAbs for tumor targeting and mechanisms of therapeutic resistance that is not only caused by the heterogeneity of tumor antigen, but also the resistance imposed by tumor microenvironment (TME), including inefficient delivery to the tumor, alteration of effector functions in the TME, and Fc-gamma receptor expression diversity and polymorphism. Further, this article provides a perspective on potential strategies to overcome these barriers and how diagnostic and prognostic biomarkers are being used in predicting response to mAb-based therapies. Overall, understanding these interdependent parameters can improve the current mAb-based formulations and develop novel mAb-based therapeutics for achieving durable clinical outcomes in a large subset of patients.
- Published
- 2021
31. Tumor microenvironment: an evil nexus promoting aggressive head and neck squamous cell carcinoma and avenue for targeted therapy
- Author
-
Nissar A. Wani, Parvaiz Yousuf, Ajaz A. Bhat, Surinder K. Batra, Shyam S. Chauhan, Mohammad Haris, Davide Bedognetti, Mushtaq A. Siddiqi, Muzafar A. Macha, Arshi Rizwan, Wael El-Rifai, and Michael P. Frenneaux
- Subjects
0301 basic medicine ,Cancer microenvironment ,Cancer Research ,Angiogenesis ,medicine.medical_treatment ,lcsh:Medicine ,Review Article ,medicine.disease_cause ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Chemoimmunotherapy ,Tumor Microenvironment ,Genetics ,medicine ,Humans ,Head and neck cancer ,lcsh:QH301-705.5 ,Tumor microenvironment ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,lcsh:R ,Correction ,medicine.disease ,Head and neck squamous-cell carcinoma ,stomatognathic diseases ,030104 developmental biology ,lcsh:Biology (General) ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer research ,business ,Carcinogenesis - Abstract
Head and neck squamous cell carcinoma (HNSCC) is a very aggressive disease with a poor prognosis for advanced-stage tumors. Recent clinical, genomic, and cellular studies have revealed the highly heterogeneous and immunosuppressive nature of HNSCC. Despite significant advances in multimodal therapeutic interventions, failure to cure and recurrence are common and account for most deaths. It is becoming increasingly apparent that tumor microenvironment (TME) plays a critical role in HNSCC tumorigenesis, promotes the evolution of aggressive tumors and resistance to therapy, and thereby adversely affects the prognosis. A complete understanding of the TME factors, together with the highly complex tumor–stromal interactions, can lead to new therapeutic interventions in HNSCC. Interestingly, different molecular and immune landscapes between HPV+ve and HPV−ve (human papillomavirus) HNSCC tumors offer new opportunities for developing individualized, targeted chemoimmunotherapy (CIT) regimen. This review highlights the current understanding of the complexity between HPV+ve and HPV−ve HNSCC TME and various tumor–stromal cross-talk modulating processes, including epithelial–mesenchymal transition (EMT), anoikis resistance, angiogenesis, immune surveillance, metastatic niche, therapeutic resistance, and development of an aggressive tumor phenotype. Furthermore, we summarize the recent developments and the rationale behind CIT strategies and their clinical applications in HPV+ve and HPV−ve HNSCC.
- Published
- 2021
32. Selective inhibition of stemness through EGFR/FOXA2/SOX9 axis reduces pancreatic cancer metastasis
- Author
-
Amar B. Singh, Quan P. Ly, Subodh M. Lele, Sanchita Rauth, Kavita Mallya, Maneesh Jain, Mokenge P. Malafa, Ramakanth Chirravuri-Venkata, Rama Krishna Nimmakayala, Raghupathy Vengoji, Parthasarathy Seshacharyulu, Garima Kaushik, Surinder K. Batra, Palanisamy Nallasamy, Moorthy P. Ponnusamy, Jason M. Foster, Satyanarayana Rachagani, and Lynette M. Smith
- Subjects
pancreatic cancer stem cells ,0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,EGFR ,Afatinib ,Biology ,Deoxycytidine ,Article ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,pancreatic cancer metastasis ,Cancer stem cell ,Cell Line, Tumor ,Pancreatic cancer ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,therapy ,Cancer ,SOX9 Transcription Factor ,medicine.disease ,Gemcitabine ,ErbB Receptors ,Pancreatic Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,medicine.drug - 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 KrasG12D/+; Pdx-1 Cre (KC) (P
- Published
- 2020
33. PAF1 cooperates with YAP1 in metaplastic ducts to promote pancreatic cancer
- Author
-
Rama Krishna Nimmakayala, Ayoola O. Ogunleye, Seema Parte, Nivedeta Krishna Kumar, Pratima Raut, Venkatesh Varadharaj, Naveen Kumar Perumal, Palanisamy Nallasamy, Sanchita Rauth, Jesse L. Cox, Subodh M. Lele, Surinder K. Batra, and Moorthy P. Ponnusamy
- Subjects
Cancer Research ,Metaplasia ,Immunology ,Pancreatic Ducts ,TEA Domain Transcription Factors ,Verteporfin ,YAP-Signaling Proteins ,Cell Biology ,Acinar Cells ,DNA-Binding Proteins ,Pancreatic Neoplasms ,Proto-Oncogene Proteins p21(ras) ,Cellular and Molecular Neuroscience ,Mice ,Cell Transformation, Neoplastic ,Animals ,Humans ,RNA Polymerase II ,Luciferases ,Ceruletide ,Carcinoma, Pancreatic Ductal ,Transcription Factors - Abstract
Acinar-to-ductal metaplasia (ADM) is a precursor lesion of pancreatic ductal adenocarcinoma (PDAC); however, the regulators of the ADM-mediated PDAC development and its targeting are poorly understood. RNA polymerase II-associated factor 1 (PAF1) maintains cancer stem cells leading to the aggressiveness of PDAC. In this study, we investigated whether PAF1 is required for the YAP1-mediated PDAC development and whether CA3 and verteporfin, small molecule inhibitors of YAP1/TEAD transcriptional activity, diminish pancreatic cancer (PC) cell growth by targeting the PAF1/YAP1 axis. Here, we demonstrated that PAF1 co-expresses and interacts with YAP1 specifically in metaplastic ducts of mouse cerulein- or KrasG12D-induced ADM and human PDAC but not in the normal pancreas. PAF1 knockdown (KD) reduced SOX9 in PC cells, and the PC cells showed elevated PAF1/YAP1 complex recruitment to the promoter of SOX9. The PAF1 KD reduced the 8xTEAD and SOX9 promoter-luciferase reporter activities in the mouse KC (KrasG12D; Pdx-1 Cre) cells and human PC cells, indicating that the PAF1 is required for the YAP1-mediated development of ADM and PC. Moreover, treatment with CA3 or verteporfin reduced the expressions of PAF1, YAP1, TEAD4, and SOX9 and decreased colony formation and stemness in KC and PC cells. CA3 treatment also reduced the viability and proliferation of PC cells and diminished the duct-like structures in KC acinar explants. CA3 or verteporfin treatment decreased the recruitment of the PAF1/YAP1 complex to the SOX9 promoter in PC cells and reduced the 8xTEAD and SOX9 promoter-luciferase reporter activities in KC and PC cells. Overall, PAF1 cooperates with YAP1 during ADM and PC development, and verteporfin and CA3 inhibit ADM and PC cell growth by targeting the PAF1/YAP1/SOX9 axis in vitro and ex vivo models. This study identified a regulatory axis of PDAC initiation and its targeting, paving the way for developing targeted therapeutic strategies for pancreatic cancer patients.
- Published
- 2022
34. Liquid biopsies to occult brain metastasis
- Author
-
Asad Ur Rehman, Parvez Khan, Shailendra Kumar Maurya, Jawed A. Siddiqui, Juan A. Santamaria-Barria, Surinder K. Batra, and Mohd Wasim Nasser
- Subjects
Cancer Research ,Oncology ,Brain Neoplasms ,Biomarkers, Tumor ,Liquid Biopsy ,Tumor Microenvironment ,Humans ,Molecular Medicine ,Breast Neoplasms ,Female ,Neoplastic Cells, Circulating - Abstract
Brain metastasis (BrM) is a major problem associated with cancer-related mortality, and currently, no specific biomarkers are available in clinical settings for early detection. Liquid biopsy is widely accepted as a non-invasive method for diagnosing cancer and other diseases. We have reviewed the evidence that shows how the molecular alterations are involved in BrM, majorly from breast cancer (BC), lung cancer (LC), and melanoma, with an inception in how they can be employed for biomarker development. We discussed genetic and epigenetic changes that influence cancer cells to breach the blood-brain barrier (BBB) and help to establish metastatic lesions in the uniquely distinct brain microenvironment. Keeping abreast with the recent breakthroughs in the context of various biomolecules detections and identifications, the circulating tumor cells (CTC), cell-free nucleotides, non-coding RNAs, secretory proteins, and metabolites can be pursued in human body fluids such as blood, serum, cerebrospinal fluid (CSF), and urine to obtain potential candidates for biomarker development. The liquid biopsy-based biomarkers can overlay with current imaging techniques to amplify the signal viable for improving the early detection and treatments of occult BrM.
- Published
- 2022
35. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments
- Author
-
Saife N. Lone, Sabah Nisar, Tariq Masoodi, Mayank Singh, Arshi Rizwan, Sheema Hashem, Wael El-Rifai, Davide Bedognetti, Surinder K. Batra, Mohammad Haris, Ajaz A. Bhat, and Muzafar A. Macha
- Subjects
Proteomics ,Cancer Research ,Oncology ,Biomarkers, Tumor ,Liquid Biopsy ,Humans ,Molecular Medicine ,Neoplastic Cells, Circulating ,Prognosis - Abstract
Over the past decade, invasive techniques for diagnosing and monitoring cancers are slowly being replaced by non-invasive methods such as liquid biopsy. Liquid biopsies have drastically revolutionized the field of clinical oncology, offering ease in tumor sampling, continuous monitoring by repeated sampling, devising personalized therapeutic regimens, and screening for therapeutic resistance. Liquid biopsies consist of isolating tumor-derived entities like circulating tumor cells, circulating tumor DNA, tumor extracellular vesicles, etc., present in the body fluids of patients with cancer, followed by an analysis of genomic and proteomic data contained within them. Methods for isolation and analysis of liquid biopsies have rapidly evolved over the past few years as described in the review, thus providing greater details about tumor characteristics such as tumor progression, tumor staging, heterogeneity, gene mutations, and clonal evolution, etc. Liquid biopsies from cancer patients have opened up newer avenues in detection and continuous monitoring, treatment based on precision medicine, and screening of markers for therapeutic resistance. Though the technology of liquid biopsies is still evolving, its non-invasive nature promises to open new eras in clinical oncology. The purpose of this review is to provide an overview of the current methodologies involved in liquid biopsies and their application in isolating tumor markers for detection, prognosis, and monitoring cancer treatment outcomes.
- Published
- 2022
36. Endothelin-axis antagonism enhances tumor perfusion in pancreatic cancer
- Author
-
Shailendra K. Gautam, Vipin Dalal, Balasrinivasa R. Sajja, Suprit Gupta, Mansi Gulati, Nidhi V. Dwivedi, Abhijit Aithal, Jesse L. Cox, Satyanarayana Rachagani, Yutong Liu, Vincent Chung, Ravi Salgia, Surinder K. Batra, and Maneesh Jain
- Subjects
Endothelin Receptor Antagonists ,Pancreatic Neoplasms ,Perfusion ,Cancer Research ,Sulfonamides ,Oncology ,Endothelins ,Tumor Microenvironment ,Humans ,Bosentan - Abstract
Delivery of therapeutic agents in pancreatic cancer (PC) is impaired due to its hypovascular and desmoplastic tumor microenvironment. The Endothelin (ET)-axis is the major regulator of vasomotor tone under physiological conditions and is highly upregulated in multiple cancers. We investigated the effect of dual endothelin receptor antagonist bosentan on perfusion and macromolecular transport in a PC cell-fibroblast co-implantation tumor model using Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI). Following bosentan treatment, the contrast enhancement ratio and wash-in rates in tumors were two- and nine times higher, respectively, compared to the controls, whereas the time to peak was significantly shorter (7.29 ± 1.29 min v/s 22.08 ± 5.88 min; p = 0.04). Importantly, these effects were tumor selective as the magnitudes of change for these parameters were much lower in muscles. Bosentan treatment also reduced desmoplasia and improved intratumoral distribution of high molecular weight FITC-dextran. Overall, these findings support that targeting the ET-axis can serve as a potential strategy to selectively enhance tumor perfusion and improve the delivery of therapeutic agents in pancreatic tumors.
- Published
- 2022
37. CXCR3 and Cognate Ligands are Associated with Immune Cell Alteration and Aggressiveness of Pancreatic Ductal Adenocarcinoma
- Author
-
Dario Ghersi, H. Carlo Maurer, Christopher M. Thompson, Surinder K. Batra, Kenneth P. Olive, Andrew Cannon, Rakesh Bhatia, Sean West, Sushil Kumar, and Pranita Atri
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Receptors, CXCR3 ,endocrine system diseases ,Microarray ,Cell ,chemical and pharmacologic phenomena ,Kaplan-Meier Estimate ,Adenocarcinoma ,Biology ,Ligands ,CXCR3 ,Chemokine CXCL9 ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,stomatognathic system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,CXCL10 ,Receptor ,Progression-Free Survival ,digestive system diseases ,Chemokine CXCL10 ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,CXCL9 ,Female ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Purpose: The cytokine milieu in pancreatic ductal adenocarcinoma (PDAC) promotes tumor progression and immune suppression, contributing to the dismal prognosis of patients with PDAC. The roles of many of these cytokines, however, have not been thoroughly investigated in PDAC. Experimental Design: PDAC microarray and The Cancer Genome Atlas datasets were analyzed to identify cytokines and cognate receptors overexpressed in PDAC and associated with survival. Pathway and CIBERSORT analyses were used to elucidate potential mechanisms of altered patient survival. Comparative analysis of cytokine expression in KPC (K-rasG12D; TP53R172H; Pdx-1cre) and KC (K-rasG12D; Pdx-1cre) PDAC models and multicolor immunofluorescence (IF) staining of human PDAC–resected samples were used to validate these findings. Results: CXCL9 and CXCL10 were among the most highly overexpressed cytokines by bioinformatics analyses, while their receptor, CXCR3, was significantly overexpressed by IHC analysis. Higher CXCR3 ligand expression was associated with shorter overall survival, while high CXCR3 expression was associated with better survival. The CXCR3 ligands, CXCL4, 9, and 10, were overexpressed in KPC compared with KC mice. Pathway analysis of CXCR3- and CXCR3 ligand–associated genes showed that CXCR3 is a marker of antitumor immunity, while its ligands may promote immunosuppression. CIBERSORT and IF studies of PDAC tissues demonstrated that high CXCR3 expression was associated with increased CD8+ T-cell and naïve B-cell signatures and loss of plasma cell signatures. CXCR3 ligand expression was associated with increased CD8+ T-cell signatures and loss of natural killer–cell signatures. Conclusions: CXCR3 ligands are overexpressed in PDAC and are associated with poor survival likely related to alterations in tumor immune infiltrate/activity.
- Published
- 2020
38. Polyanhydride nanoparticles stabilize pancreatic cancer antigen <scp>MUC4β</scp>
- Author
-
Luman Liu, Joyce C. Solheim, Sushil Kumar, John Christiansen, Michael J. Wannemuehler, Mansi Gulati, Shailendra K. Gautam, Prakash Kshirsagar, Balaji Narasimhan, Surinder K. Batra, Maneesh Jain, and Abhijit Aithal
- Subjects
Antigenicity ,Materials science ,medicine.drug_class ,medicine.medical_treatment ,0206 medical engineering ,Biomedical Engineering ,02 engineering and technology ,Protein degradation ,Monoclonal antibody ,Cancer Vaccines ,Article ,Epitope ,Biomaterials ,Epitopes ,Mice ,Immune system ,Antigen ,Antigens, Neoplasm ,Polyanhydrides ,medicine ,Animals ,Humans ,Drug Carriers ,Mucin-4 ,Immunogenicity ,Metals and Alloys ,Immunotherapy ,021001 nanoscience & nanotechnology ,020601 biomedical engineering ,Pancreatic Neoplasms ,Ceramics and Composites ,Cancer research ,Nanoparticles ,0210 nano-technology - Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies and represents an increasing and challenging threat, especially with an aging population. The identification of immunogenic PC-specific upregulated antigens and an enhanced understanding of the immunosuppressive tumor microenvironment have provided opportunities to enable the immune system to recognize cancer cells. Due to its differential upregulation and functional role in PC, the transmembrane mucin MUC4 is an attractive target for immunotherapy. In the current study we characterized the antigen stability, antigenicity and release kinetics of a MUC4β-nanovaccine to guide further optimization and, in vivo evaluation. Amphiphilic polyanhydride copolymers based on 20 mol % 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane and 80 mol % 1,6-bis(p-carboxyphenoxy)hexane were used to synthesize nanoparticles. Structurally stable MUC4β protein was released from the particles in a sustained manner and characterized by gel electrophoresis and fluorescence spectroscopy. Modest levels of protein degradation were observed upon release. The released protein was also analyzed by MUC4β-specific monoclonal antibodies using ELISA and showed no significant loss of epitope availability. Further, mice immunized with multiple formulations of combination vaccines containing MUC4β-loaded nanoparticles generated MUC4β-specific antibody responses. These results indicate that polyanhydride nanoparticles are viable MUC4β vaccine carriers, laying the foundation for evaluation of this platform for PC immunotherapy.
- Published
- 2020
39. Unraveling mucin domains in cancer and metastasis: when protectors become predators
- Author
-
Saravanakumar Marimuthu, Sushil Kumar, Koelina Ganguly, Sanchita Rauth, and Surinder K. Batra
- Subjects
0301 basic medicine ,Cancer Research ,Inflammation ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Neoplasms ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,MUC1 ,Tissue homeostasis ,chemistry.chemical_classification ,Agrin ,Mucin ,Mucins ,Transmembrane protein ,Cell biology ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,medicine.symptom ,Carcinogenesis ,Glycoprotein - Abstract
A dynamic mucosal layer shields the epithelial cells lining the body cavities and is made up of high molecular weight, heavily glycosylated, multidomain proteins called mucins. Mucins, broadly grouped into transmembrane and secreted mucins, are the first responders to any mechanical or chemical insult to the epithelia and help maintain tissue homeostasis. However, their intrinsic properties to protect and repair the epithelia are exploited during oncogenic processes, where mucins are metamorphosed to aid the tumor cells in their malignant journey. Diverse domains, like the variable number tandem repeats (VNTR), sea urchin sperm protein enterokinase and agrin (SEA), adhesion-associated domain (AMOP), nidogen-like domain (NIDO), epidermal growth factor-like domain (EGF), and von Willebrand factor type D domain (vWD) on mucins, including MUC1, MUC4, MUC5AC, MUC5B, and MUC16, have been shown to facilitate cell-to-cell and cell-to-matrix interactions, and cell-autonomous signaling to promote tumorigenesis and distant dissemination of tumor cells. Several obstacles have limited the study of mucins, including technical difficulties in working with these huge glycoproteins, the dearth of scientific tools, and lack of animal models; thus, the tissue-dependent and domain-specific roles of mucins during mucosal protection, chronic inflammation, tumorigenesis, and hematological dissemination of malignant cells are still unclear. Future studies should try to integrate information on the rheological, molecular, and biological characteristics of mucins to comprehensively delineate their pathophysiological role and evaluate their suitability as targets in future diagnostic and therapeutic strategies.
- Published
- 2020
40. Targeting the IκB Kinase Enhancer and Its Feedback Circuit in Pancreatic Cancer☆
- Author
-
Jin Q. Cheng, Domenico Coppola, Surinder K. Batra, Richard B. Kim, Sridevi Challa, Kazim Husain, and Mokenge P. Malafa
- Subjects
0301 basic medicine ,Trametinib ,MAPK/ERK pathway ,Cancer Research ,Original article ,business.industry ,MEK inhibitor ,IκB kinase ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cancer stem cell ,030220 oncology & carcinogenesis ,Pancreatic cancer ,Cancer research ,medicine ,IKBKE ,ERBB3 ,business - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with an overall median 5-year survival rate of 8%. This poor prognosis is because of the development of resistance to chemotherapy and radiation therapy and lack of effective targeted therapies. IκB kinase enhancer (IKBKE) overexpression was previously implicated in chemoresistance. Because IKBKE is frequently elevated in PDAC and IKBKE inhibitors are currently in clinical trials, we evaluated IKBKE as a therapeutic target in this disease. Depletion of IKBKE was found to significantly reduce PDAC cell survival, growth, cancer stem cell renewal, and cell migration and invasion. Notably, IKBKE inhibitor CYT387 and IKBKE knockdown dramatically activated the MAPK pathway. Phospho-RTK array analyses showed that IKBKE inhibition leads to rapid upregulation of ErbB3 and IGF-1R expression, which results in MAPK-ERK pathway activation-thereby limiting the efficacy of IKBKE inhibitors. Furthermore, IKBKE inhibition leads to stabilization of FOXO3a, which is required for RTK upregulation on IKBKE inhibition. Finally, we demonstrated that the IKBKE inhibitors synergize with the MEK inhibitor trametinib to significantly induce cell death and inhibit tumor growth and liver metastasis in an orthotopic PDAC mouse model.
- Published
- 2020
41. Delivery of radioimmunotherapy for solid tumors
- Author
-
Shailendra K. Gautam, Vipin Dalal, Maneesh Jain, and Surinder K. Batra
- Subjects
Tumor microenvironment ,business.industry ,medicine.drug_class ,Core component ,medicine.medical_treatment ,Vascular transport ,Monoclonal antibody ,Clinical success ,Conventional radiotherapy ,Stroma ,Radioimmunotherapy ,medicine ,Cancer research ,business - Abstract
The clinical success of radioimmunotherapy (RIT) in hematological malignancies could not be recapitulated to treat solid tumors. The delivery of macromolecule-based RIT agents in solid tumors is challenging due to a complex tumor microenvironment that includes disrupted vascular transport and stroma induced physical and biochemical impediments. The inter- and intratumoral heterogeneities within and across the solid malignancies result in disproportionate delivery of radioimmunoconjugates (RICs). As the tumor-specific monoclonal antibodies and therapeutic radionuclides are two core components of RIT, several efforts have been directed to optimize the size, stability, and delivery of RICs to develop clinically relevant alternatives of conventional radiotherapy. Nevertheless, recent trends are more focused on combining RIT with chemotherapies, immunotherapies, antistromal therapies, and antivascular therapies to enhance tumor-specific delivery and therapeutic efficacy. In this chapter, we discuss the major challenges and recent efforts to improve the effectiveness of RIT in solid tumors.
- Published
- 2022
42. Clinical and Molecular Attributes and Evaluation of Pancreatic Cystic Neoplasm
- Author
-
Pratima, Raut, Rama Krishna, Nimmakayala, Surinder K, Batra, and Moorthy P, Ponnusamy
- Subjects
Cancer Research ,Oncology ,Genetics - Abstract
Intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs) are all considered "Pancreatic cystic neoplasms (PCNs)" and show a varying risk of developing into pancreatic ductal adenocarcinoma (PDAC). These lesions display different molecular characteristics, mutations, and clinical manifestations. A lack of detailed understanding of PCN subtype characteristics and their molecular mechanisms limits the development of efficient diagnostic tools and therapeutic strategies for these lesions. Proper in vivo mouse models that mimic human PCNs are also needed to study the molecular mechanisms and for therapeutic testing. A comprehensive understanding of the current status of PCN biology, mechanisms, current diagnostic methods, and therapies will help in the early detection and proper management of patients with these lesions and PDAC. This review aims to describe all these aspects of PCNs, specifically IPMNs, by describing the future perspectives.
- Published
- 2023
43. Role of Neuropilin-2-mediated signaling axis in cancer progression and therapy resistance
- Author
-
Ridwan Islam, Juhi Mishra, Sanika Bodas, Sreyashi Bhattacharya, Surinder K. Batra, Samikshan Dutta, and Kaustubh Datta
- Subjects
Cancer Research ,Oncology ,Neovascularization, Pathologic ,Neoplasms ,Tumor Microenvironment ,Humans ,Neuropilins ,Neuropilin-2 ,Signal Transduction - Abstract
Neuropilins (NRPs) are transmembrane proteins involved in vascular and nervous system development by regulating angiogenesis and axon guidance cues. Several published reports have established their role in tumorigenesis. NRPs are detectable in tumor cells of several cancer types and participate in cancer progression. NRP2 is also expressed in endothelial and immune cells in the tumor microenvironment and promotes functions such as lymphangiogenesis and immune suppression important for cancer progression. In this review, we have taken a comprehensive approach to discussing various aspects of NRP2-signaling in cancer, including its regulation, functional significance in cancer progression, and how we could utilize our current knowledge to advance the studies and target NRP2 to develop effective cancer therapies.
- Published
- 2021
44. MARK2 regulates chemotherapeutic responses through class IIa HDAC-YAP axis in pancreatic cancer
- Author
-
Yongji Zeng, Ling Yin, Jiuli Zhou, Renya Zeng, Yi Xiao, Adrian R. Black, Tuo Hu, Pankaj K. Singh, Feng Yin, Surinder K. Batra, Fang Yu, Yuanhong Chen, and Jixin Dong
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Paclitaxel ,Cell Line, Tumor ,Genetics ,Animals ,Mitosis ,Molecular Biology ,Histone Deacetylases ,Article - Abstract
Despite paclitaxel’s wide use in cancer treatment, patient response rate is still low and drug resistance is a major clinical obstacle. Through a Phos-tag-based kinome-wide screen, we identified MARK2 as a critical regulator for paclitaxel chemosensitivity in PDAC. We show that MARK2 is phosphorylated by CDK1 in response to antitubulin chemotherapeutics and in unperturbed mitosis. Phosphorylation is essential for MARK2 in regulating mitotic progression and paclitaxel cytotoxicity in PDAC cells. Mechanistically, our findings also suggest that MARK2 controls paclitaxel chemosensitivity by regulating class IIa HDACs. MARK2 directly phosphorylates HDAC4 specifically during antitubulin treatment. Phosphorylated HDAC4 promotes YAP activation and controls expression of YAP target genes induced by paclitaxel. Importantly, combination of HDAC inhibition and paclitaxel overcomes chemoresistance in organoid culture and preclinical PDAC animal models. The expression levels of MARK2, HDACs, and YAP are upregulated and positively correlated in PDAC patients. Inhibition of MARK2 or class IIa HDACs potentiates paclitaxel cytotoxicity by inducing mitotic abnormalities in PDAC cells. Together, our findings identify the MARK2-HDAC axis as a druggable target for overcoming chemoresistance in PDAC.
- Published
- 2021
45. MiR-212-3p functions as a tumor suppressor gene in group 3 medulloblastoma via targeting nuclear factor I/B (NFIB)
- Author
-
Naveenkumar Perumal, Ranjana K. Kanchan, David Doss, Noah Bastola, Pranita Atri, Ramakanth C. Venkata, Ishwor Thapa, Raghupathy Vengoji, Shailendra K. Maurya, David Klinkebiel, Geoffrey A. Talmon, Mohd W. Nasser, Surinder K. Batra, and Sidharth Mahapatra
- Subjects
Homeobox protein NANOG ,Tumor suppressor gene ,Biology ,Metastasis ,Pathology and Forensic Medicine ,miR-212-3p ,Mice ,Cellular and Molecular Neuroscience ,SOX2 ,medicine ,Animals ,Humans ,Gene silencing ,Cerebellar Neoplasms ,RC346-429 ,Cells, Cultured ,Research ,Wnt signaling pathway ,Cell cycle ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,MicroRNAs ,NFI Transcription Factors ,Group 3 medulloblastoma ,c-Myc ,NFIB ,Nuclear factor I/B ,Cancer research ,17p13.3 ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,Medulloblastoma - Abstract
BackgroundMedulloblastoma (MB), the most frequent malignant pediatric brain tumor, is subdivided into four primary subgroups, wingless-type (WNT), sonic hedgehog (SHH), group 3, and group 4. Haploinsufficiency of chromosome 17p13.3 and c-Myc amplification distinguish high-risk group 3 tumors associated with rapid metastasis, recurrence and early mortality. We sought to identify the role of miR-212-3p, which resides on chromosome 17p13.3, in the pathophysiology of group 3 MB.MethodsWe first determined miR-212-3p expression in group 3 MB using several publicly-available datasets with confirmatory studies in vitro. We then identified epigenetic regulation by studying methylation and HDAC modifications along the promoter region. We used two systems for expression restoration, i.e. transient transfection or stable induction, to delineate miR-212-3p’s tumor suppressive and biochemical properties via assays assessing cancer proliferation, migration, invasion, colony formation, along with cell cycle and apoptosis analyses. We then compared MB and miR target databases to isolate a putative target whose biochemical and oncogenic properties were similarly elucidated using either transient silencing of target expression or stable induction of miR-212-3p.ResultsRNA expression analyses revealed dramatically reduced miR-212-3p levels in group 3 tumors and cell lines mainly through epigenetic silencing via histone modifications. Restoring miR-212-3p expression reduced in vitro cancer cell proliferation, migration, colony formation, and wound healing. Elevated miR-212-3p levels shifted c-Myc phosphorylation (from serine-62 to threonine-58), triggering destabilization and degradation; concurrently, its pro-apoptotic binding partners, i.e., Bin-1 and P19ARF, were upregulated with subsequent elevated apoptotic signals. Using a combination of transcriptomic data and dual luciferase assay, we isolated an oncogenic target of miR-212-3p, i.e. NFIB, a nuclear transcription factor implicated in metastasis and recurrence in various cancers. Increased expression of NFIB was confirmed in group 3 tumors, with poor survival shown in high-expressing patients. Transient NFIB silencing in vitro reduced cancer cell proliferation, colony formation, migration, and invasion. Concurrently, in group 3 MB cells, reduced medullosphere formation along with decreased expression of stem cell markers (Nanog, Oct4, Sox2, CD133) were noted.ConclusionThese results substantiate the tumor-suppressive role of miR-212-3p in group 3 MB and provide a potential therapeutic oncogenic target implicated in metastasis and tumor recurrence.
- Published
- 2021
46. Correction: Kaushal et al. Repurposing Niclosamide for Targeting Pancreatic Cancer by Inhibiting Hh/Gli Non-Canonical Axis of Gsk3β. Cancers 2021, 13, 3105
- Author
-
Surya K. Mallapragada, Rakesh Bhatia, Jyoti B. Kaushal, Quan P. Ly, Surinder K. Batra, Satyanarayana Rachagani, Pratima Raut, and Ranjana Kanchan
- Subjects
Cancer Research ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Correction ,medicine.disease ,n/a ,Oncology ,Non canonical ,Pancreatic cancer ,medicine ,Cancer research ,business ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,RC254-282 ,Niclosamide ,Repurposing ,medicine.drug - Abstract
Niclosamide (Nic), an FDA-approved anthelmintic drug, is reported to have anti-cancer efficacy and is being assessed in clinical trials for various solid tumors. Based on its ability to target multiple signaling pathways, in the present study, we evaluated the therapeutic efficacy of Nic on pancreatic cancer (PC) in vitro. We observed an anti-cancerous effect of this drug as shown by the G0/G1 phase cell cycle arrest, inhibition of PC cell viability, colony formation, and migration. Our results revealed the involvement of mitochondrial stress and mTORC1-dependent autophagy as the predominant players of Nic-induced PC cell death. Significant reduction of Nic-induced reactive oxygen species (ROS) and cell death in the presence of a selective autophagy inhibitor spautin-1 demonstrated autophagy as a major contributor to Nic-mediated cell death. Mechanistically, Nic inhibited the interaction between BCL2 and Beclin-1 that supported the crosstalk of autophagy and apoptosis. Further, Nic treatment resulted in Gsk3β inactivation by phosphorylating its Ser-9 residue leading to upregulation of Sufu and Gli3, thereby negatively impacting hedgehog signaling and cell survival. Nic induced autophagic cell death, and p-Gsk3b mediated Sufu/Gli3 cascade was further confirmed by Gsk3β activator, LY-294002, by rescuing inactivation of Hh signaling upon Nic treatment. These results suggested the involvement of a non-canonical mechanism of Hh signaling, where p-Gsk3β acts as a negative regulator of Hh/Gli1 cascade and a positive regulator of autophagy-mediated cell death. Overall, this study established the therapeutic efficacy of Nic for PC by targeting p-Gsk3β mediated non-canonical Hh signaling and promoting mTORC1-dependent autophagy and cell death.
- Published
- 2021
47. Differential gene expression-based connectivity mapping identified novel drug candidate and improved Temozolomide efficacy for Glioblastoma
- Author
-
Raghupathy Vengoji, Pranita Atri, Moorthy P. Ponnusamy, Parthasarathy Seshacharyulu, Satyanarayana Rachagani, Lynette M. Smith, Muzafar A. Macha, Yutong Liu, Maneesh Jain, Sidharth Mahapatra, Surinder K. Batra, Naveenkumar Perumal, Kavita Mallya, and Nicole Shonka
- Subjects
Cancer Research ,Methyltransferase ,Cell Survival ,Abexinostat ,Mice, Transgenic ,In silico analysis ,Mice ,chemistry.chemical_compound ,HDAC ,Cell Line, Tumor ,Drug Discovery ,Temozolomide ,medicine ,Animals ,Humans ,Connectivity map ,Antineoplastic Agents, Alkylating ,Vorinostat ,RC254-282 ,Blood-brain barrier ,Dose-Response Relationship, Drug ,HDAC11 ,business.industry ,Histone deacetylase 2 ,Gene Expression Profiling ,Research ,DNA Breaks ,Computational Biology ,Disease Management ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xenograft Model Antitumor Assays ,HDAC1 ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Treatment Outcome ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Histone deacetylase ,Transcriptome ,Glioblastoma ,business ,medicine.drug - 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 - PTENflox/+; 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.
- Published
- 2021
48. Acinar to ductal cell trans-differentiation: A prelude to dysplasia and pancreatic ductal adenocarcinoma
- Author
-
Seema Parte, Rama Krishna Nimmakayala, Surinder K. Batra, and Moorthy P. Ponnusamy
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Metaplasia ,Cell Transformation, Neoplastic ,Oncology ,Cell Transdifferentiation ,Genetics ,Humans ,Acinar Cells ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic cancer (PC) is the deadliest neoplastic epithelial malignancies and is projected to be the second leading cause of cancer-related mortality by 2024. Five years overall survival being ~10%, mortality and incidence rates are disturbing. Acinar to ductal cell metaplasia (ADM) encompasses cellular reprogramming and phenotypic switch-over, making it a cardinal event in tumor initiation. Differential cues and varied regulatory factors drive synchronous functions of metaplastic cell populations leading to multiple cell fates and physiological outcomes. ADM is a precursor for developing early pre-neoplastic lesions further progressing into PC due to oncogenic signaling. Hence delineating molecular events guiding tumor initiation may provide cues for regenerative medicine and precision onco-medicine. Therefore, understanding PC pathogenesis and early diagnosis are crucial. We hereby provide a timely overview of the current progress in this direction and future perspectives we foresee unfolding in the best interest of patient well-being and better clinical management of PC.
- Published
- 2021
49. Emerging Role of miR-345 and Its Effective Delivery as a Potential Therapeutic Candidate in Pancreatic Cancer and Other Cancers
- Author
-
Maia M. C. Bennett, Brianna M. White, Nagabhishek Sirpu Natesh, Rakhee Rathnam Kalari Kandy, Surya K. Mallapragada, Metin Uz, Satyanarayana Rachagani, and Surinder K. Batra
- Subjects
Pancreatic ductal adenocarcinoma ,business.industry ,Mechanism (biology) ,Drug discovery ,Pharmaceutical Science ,PDAC ,Review ,miR-345 ,medicine.disease ,Malignancy ,Metastasis ,RS1-441 ,Pharmacy and materia medica ,Downregulation and upregulation ,Pancreatic cancer ,microRNA ,Cancer research ,Medicine ,business ,miRNA nanodelivery - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with high mortality, poor prognosis, and palliative treatments, due to the rapid upregulation of alternative compensatory pathways and desmoplastic reaction. miRNAs, small non-coding RNAs, have been recently identified as key players regulating cancer pathogenesis. Dysregulated miRNAs are associated with molecular pathways involved in tumor development, metastasis, and chemoresistance in PDAC, as well as other cancers. Targeted treatment strategies that alter miRNA levels in cancers have promising potential as therapeutic interventions. miRNA-345 (miR-345) plays a critical role in tumor suppression and is differentially expressed in various cancers, including pancreatic cancer (PC). The underlying mechanism(s) and delivery strategies of miR-345 have been investigated by us previously. Here, we summarize the potential therapeutic roles of miR-345 in different cancers, with emphasis on PDAC, for miRNA drug discovery, development, status, and implications. Further, we focus on miRNA nanodelivery system(s), based on different materials and nanoformulations, specifically for the delivery of miR-345.
- Published
- 2021
50. Contribution of CXCR3-Mediated Signaling in the Metastatic Cascade of Solid Malignancies
- Author
-
Rakhee Rathnam Kalari Kandy, Andrew Cannon, Christopher M. Thompson, Joyce C. Solheim, Surinder K. Batra, Sushil Kumar, and Rakesh Bhatia
- Subjects
Cancer Research ,Tumor microenvironment ,Receptors, CXCR3 ,business.industry ,Cancer ,chemical and pharmacologic phenomena ,CXCR3 ,medicine.disease ,Phenotype ,Article ,Metastasis ,stomatognathic diseases ,Chemokine receptor ,Immune system ,stomatognathic system ,Oncology ,Neoplasms ,Cancer cell ,Genetics ,Cancer research ,Medicine ,Humans ,Neoplasm Metastasis ,business ,Signal Transduction - Abstract
Metastasis is a significant cause of the mortality resulting from solid malignancies. The process of metastasis is complex and is regulated by numerous cancer cell-intrinsic and - extrinsic factors. CXCR3 is a chemokine receptor that is frequently expressed by cancer cells, endothelial cells and immune cells. CXCR3A signaling in cancer cells tends to promote the invasive and migratory phenotype of cancer cells. Indirectly, CXCR3 modulates the anti-tumor immune response resulting in variable effects that can permit or inhibit metastatic progression. Finally, the activity of CXCR3B in endothelial cells is generally angiostatic, which limits the access of cancer cells to key conduits to secondary sites. However, the interaction of these activities within a tumor and the presence of opposing CXCR3 splice variants clouds the picture of the role of CXCR3 in metastasis. Consequently, thorough analysis of the contributions of CXCR3 to cancer metastasis is necessary. This review is an in-depth examination of the involvement of CXCR3 in the metastatic process of solid malignancies.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.