GET THE APP

Molecular Oncogenesis of Intraductal Papillary Mucinous Neoplasms of the Pancreas and its Possible Molecular Targeted Therapy
..

Molecular and Genetic Medicine

ISSN: 1747-0862

Open Access

Review Article - (2023) Volume 17, Issue 2

Molecular Oncogenesis of Intraductal Papillary Mucinous Neoplasms of the Pancreas and its Possible Molecular Targeted Therapy

Mitsuru Sakitani*
*Correspondence: Mitsuru Sakitani, Director of the Institute CCC, Kobe, Hyogo, Japan, Email:
Director of the Institute CCC, Kobe, Hyogo, Japan

Received: 01-Mar-2023, Manuscript No. jmgm-23-92176; Editor assigned: 03-Mar-2023, Pre QC No. P-92176; Reviewed: 17-Mar-2023, QC No. Q-92176; Revised: 23-Mar-2023, Manuscript No. R-92176; Published: 31-Mar-2023 , DOI: 10.37421/1747-0862.2023.17.602
Citation: Sakitani, Mitsuru. “Molecular Oncogenesis of Intraductal Papillary Mucinous Neoplasm of the Pancreas and its Possible Molecular Targeted Therapy.” J Mol Genet Med 17 (2023): 602
Copyright: © 2023 Sakitani M. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Intraductal papillary neoplasm of the pancreas (IPMN) is a frequently found, pancreatic cystic neoplasm. IPMN has relatively high malignant potential, and its therapeutic strategy is limited to surgical resection. It is well known that mutations of GNAS and KRAS play important roles in its malignant progression, but its molecular mechanisms have not been well elucidated. In this review, clinical features and molecular alterations of IPMN were summarized. Then, crosstalk between KRAS signaling and phosphatidylinositol 3-kinase (PI3K) signaling was clarified. Finally, it was indicated that the final effector of KRAS mutant IPMN could be carbon anhydrase IX (CA9), and the possibility of molecular targeted therapy against IPMN by means of CA9 inhibitors was discussed.

Keywords

IPMN • PDAC • GNAS • KRAS • PI3K • HIF • CA9

Introduction

Intraductal papillary neoplasm of the pancreas (IPMN) [1-3] is a frequently found, pancreatic cystic neoplasm [4-6]. IPMN has relatively high malignant potential [7]. Thus its malignant progression causes serious problems in clinical practice [3,8]. At the overt malignant stage, invasive IPMN shows poor prognosis comparable to that of pancreatic ductal adenocarcinoma (PDAC) [9]. It is well known that mutations of GNAS and KRAS play important roles in its malignant progression [10-12], but its molecular mechanisms have not been well elucidated. In addition, there is no effective cytotoxic chemotherapy nor molecular targeted therapy [11]. Instead, surgical resection of pancreatic lesions is only the current recommended treatment strategy [1-3,13,14], and the effect of adjuvant chemotherapy after surgical operation is quite limited [15,16] or questioned [11,17].

In this regard, a recent indication of a novel therapy against KRAS mutant IPMN by means of inhibitors of carbon anhydrase IX (CA9) [18] is worth noting. In this review, we summarize clinical features of IPMN and molecular alterations, in particular GNAS and KRAS mutations. Then, we clarify crosstalk between KRAS signaling and phosphatidylinositol 3-kinase (PI3K) signaling via direct binding of KRAS to PI3K and via hypoxia inducible factor (HIF). Finally, we indicate that one of the final effector of KRAS mutant IPMN can be CA9 and discuss the possible molecular targeted therapy against IPMN by CA9 inhibitors.

Discussion

Clinical features of IPMN

IPMN is classified into three clinical types, i.e., branch duct IPMN (BDIBDIPMN), main duct IPMN (MD-IPMN), and mixed type IPMN [1,2]. Prognosis of BD-IPMN is better than that of MD-IPMN or mixed type IPMN [2,9,19]. Histological classification of IPMN [2] is grouped into gastric type with lowgrade dysplasia (LGD), intestinal type with high-grade dysplasia (HGD), and pancreatobiliary type with HGD [3,11]. The rare oncocytic type was also known [3], but this is now reclassified into intraductal oncocytic papillary neoplasm [12]. Invasive cancer associated with IPMN shows tubular type or colloid type [3,11]. Thus the histological malignant progression of IPMN is summarized as from LGD (gastric type) through HGD (intestinal or pancreatobiliary types) to invasive IPMN (tubular or colloid types) as in Table 1.

Table 1:Malignant progression and histological types of IPMN.

Grade Frequent histological types
Low-grade dysplasia Gastric type
High-grade dysplasia Intestinal or pancreatobiliary types
Invasive cancer associated with IPMN Tiubular or colloid types

Molecular alterations of IPMN

There are several gene mutations of IPMN [10-12,20-23]. Among them, mutations of KRAS [24,25] and GNAS [26,27] are thought to play crucial roles in oncogenesis of IPMN [11,12]. More than 90% of IPMN harbor KRAS and/ or GNAS mutations [20]. KRAS mutation is detected in up to 80% of IPMN [10,11,21] and GNAS mutations is found in around 70% of IPMN [10,21], while coexistence of both KRAS and GNAS mutations is observed in more than 30% of IPMN cases [11,12,20].

KRAS mutation (G12D and G12V are frequent mutation sites) [28-30] activates downstream effectors including RAF, mitogen-activated protein kinase kinase (MEK), and extra-cellular signal-regulated kinase (ERK) [31] in Figure 1. The KRAS/RAF/MEK/ERK signaling further activates transcription of various target genes [32-34] and translation of HIF mRNA [35-38]. KRAS also activates PI3K by direct binding [39-41]. Crosstalk between KRAS signaling and PI3K signaling will be later discussed.

GNAS encodes G-protein stimulatory α subunit (Gsα), GNAS has hotspots of activating mutation H201H, H201C and Q227L [11,42], and mutant GNAS disrupts GTPase activity via structural change in the GTPase domain of Gsα, leading to constitutive activation of downstream signaling pathways [42] such as cAMP and cAMP-dependent protein kinase A (PKA) [42-44]. This cAMPPKA signaling is involved in oncogenesis of many cancer [45] in Figure 1. In IPMN and PDAC, suppression of tumor suppressor salt inducible kinase (SIK) by GNAS signaling is supposed to be critical to oncogenesis [43]. In addition, mutations of RNF4 (25-75%), TP53 (18-20%), SMAD4 (<5%), KLF5 (15%) and CDKN2A (0-15%) are found in HGD of IPMN [10]. Their roles in malignant progression have been under intensive investigation [11,28,43,46-48].

Crosstalk between KRAS/MEK/ERK and PI3K/AKT/mTOR signaling via PI3K

The KRAS/MEK/ERK signaling has crosstalk with the PI3K/AKT/ mechanistic target of rapamycin (mTOR) signaling pathway (Figure 1 and Table 2). This crosstalk has been overlooked but has critical importance in elucidating oncogenesis of IPMN and developing novel molecular targeted therapies against IPMN.

Figure

Figure 1.KRAS and GNAS signaling pathways, and crosstalk between KRAS and PI3K signaling pathways. Abbreviation: CA9, carbon anhydrase IX; Gsα, G-protein stimulatory α subunit; HIF, hypoxia inducible factor; mTOR, mechanistic target of rapamycin; PI3K, phosphatidylinositol 3-kinase; PKA, protein kinase A; SIK, salt inducible kinase.

Table 2:Crosstalk between KRAS/MER/ERK and PI3K/AKT/mTOR signaling pathways.

Target Mechanism
PI3K Direct binding of KRAS
Translation of HIF-1α mRNA Activation of eIFs by ERK1/ERK2 or mTORC1

First, crosstalk between KRAS and PI3K is induced by direct binding of KRA to PI3K [39-41]. PI3Kα has RAS-binding domain of p110α [49-51]. After activated by binding of KRAS with G12D or G12V mutations, the KRAS-PI3K complex further activates its downstream effectors AKT (protein kinase B) and mTOR complex 1 (mTORC1) [40,41].

Crosstalk between KRAS/MEK/ERK and PI3K/AKT/mTOR signaling via HIF

Second, crosstalk between the KRAS/MEK/ERK and the PI3K/AKT/mTOR signaling is due to the common activation pathway via translation activation of HIF-1α mRNA [18,35-38]. Activated mTORC1 stimulates phosphorylation of 4E-binding proteins (4E-BP1), which induces its release from eIF4E, leading to association of eIF4E with eIF4G. This then induces assembly and activation of the mRNA-cap binding eIF4F complex (eIF4E, eIF4G and eIF4A) [52-54]. The activated eIF4F complex stimulates translation of mRNA of HIF-1α. In addition, mTORC1 activates ribosomal S6 kinases (S6Ks).

The relevant substrates of S6Ks in translation regulation are ribosomal protein S6 (rpS6), eIF4B, eukaryotic elongation factor 2 kinase (eEF2K) and programmed cell death 4 protein (PDCD4) [54], which finally stimulate translation of mRNA of HIF-1α. Activated ERK1 or ERK2 phosphorylates 4E-BP1, S6K, and MAP kinase interacting kinase (MNK) [36,55]. MNK also phosphorylates eIF-4E. ERK also increases HIF-1α activity by phosphorylation of coactivator CBP/p300 [55]. Taken together, crosstalk between the KRAS/MEK/ERK and the PI3K/AKT/mTOR signaling is mediated by HIF-1α signaling via activation of its translation.

CA9 the final effector

The final effector of the mutant KRAS is supposed to be CA9. First, HIF-1α is commonly stimulated by both the KRAS/MEK/ERK and PI3K/AKT/mTOR signaling pathways. Then, CA9 is activated by HIF-1α via hypoxia response element (HRE) in its promoter region with recruitment of CBP/p300 [56-58]. Thus, the final effector of the KRAS mutant IPMN is suspected to be CA9. In fact,CA9 has multiple functions to promote oncogenesis in many cancers by cell proliferation via correction of intracellular pHi and extracellular pHe [59,60], cell survival via apoptosis inhibition [61,62], promotion of metastasis via cell migration and invasion [63-65], therapy resistance via phenotypic plasticity [66-68], and tumor expansion via tumorigenicity [69].

Second, molecular targeted therapies against KRAS mutant PDAC by means of inhibitors against effectors in the KRAS/MEK/ERK signaling pathway are ineffective [70,71], but inhibitors of CA9 effectively prevent cell growth of KRAS mutant PDAC cell lines [31,72,73]. These results indicate not only effectivity of CA9 inhibitors against KRAS mutant PDAC but also the possibility of novel therapies against KRAS mutant IPMN by means of CA9 inhibitors.

Conclusion

The mutations of GNAS and KRAS are critical to oncogenesis of IPMN. In the GNAS signaling, SIK is suspected to play an essential role, but its mechanisms should be further clarified. On the contrary, in the KRAS signaling, both the KRAS/ERK/HIF/CA9 axis and the PI3K/mTORC1/HIF/CA9 axis commonly activate the final effector CA9, and CA9 inhibitors are suspected to be effective against KRAS mutant PDAC. As a logical consequence, in KRAS mutant IPMN, CA9 can be involved in oncogenesis, and CA9 inhibitors against KRAS mutant IPMN are expected to be a possible therapeutic strategy. Further investigation will be required.

Conflict of Interest

The author declares that there is no conflict of interests.

References

  1. Tanaka, Masao, Suresh Chari, Volkan Adsay and Fernandez-Del Carlos Castillo, et al. "International consensus guidelines for management of intraductal papillary mucinous neoplasms and mucinous cystic neoplasms of the pancreas." Pancreatology 6 (2006): 17-32.
  2. Google Scholar, Crossref, Indexed at

  3. Tanaka, Masao, Carlos Fernández-del Castillo, Volkan Adsay and Suresh Chari, et al. "International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas." Pancreatology 12 (2012): 183-197.
  4. Google Scholar, Crossref, Indexed at

  5. Tanaka, Masao, Carlos Fernández-del Castillo, Terumi Kamisawa and Jin Young Jang, et al. "Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas." Pancreatology 17 (2017): 738-753.
  6. Google Scholar, Crossref, Indexed at

  7. Reid-Lombardo, Kaye M., Jennifer St Sauver, Zhuo Li and William A. Ahrens, et al. "Incidence, prevalence, and management of intraductal papillary mucinous neoplasm in olmsted county, minnesota, 1984–2005 a population study." Pancreas 37 (2008): 139.
  8. Google Scholar, Crossref, Indexed at

  9. Ferrone, Cristina R., Camilo Correa-Gallego, Andrew L. Warshaw and William R. Brugge, et al. "Current trends in pancreatic cystic neoplasms." Arch Surg 144 (2009): 448-454.
  10. Google Scholar, Crossref, Indexed at

  11. Chen, Wei, Nehaal Ahmed and Somashekar G. Krishna. "Pancreatic cystic lesions: A focused review on cyst clinicopathological features and advanced diagnostics." Diagnostics 13 (2023): 65.
  12. Google Scholar, Crossref Indexed at

  13. Tanaka, Masao. "Intraductal papillary mucinous neoplasm of the pancreas as the main focus for early detection of pancreatic adenocarcinoma." Pancreas 47 (2018): 544-550.
  14. Google Scholar, Crossref Indexed at

  15. Vege, Santhi Swaroop, Barry Ziring, Rajeev Jain and Paul Moayyedi, et al. "American gastroenterological association institute guideline on the diagnosis and management of asymptomatic neoplastic pancreatic cysts." Gastroenterol 148 (2015): 819-822.
  16. Google Scholar, Crossref Indexed at

  17. Schnelldorfer, Thomas, Michael G. Sarr, David M. Nagorney and Lizhi Zhang, et al. "Experience with 208 resections for intraductal papillary mucinous neoplasm of the pancreas." Arch Surg 143 (2008): 639-646.
  18. Google Scholar, Crossref Indexed at

  19. Nasca, Vincenzo, Marta Chiaravalli, Geny Piro and Annachiara Esposito, et al. "Intraductal pancreatic mucinous neoplasms: A tumor-biology based approach for risk stratification." Int J Mol Sci 21 (2020): 6386.
  20. Google Scholar, Crossref Indexed at

  21. Mas, Léo, Renato M. Lupinacci, Jérôme Cros and Jean-Baptiste Bachet, et al. "Intraductal papillary mucinous carcinoma versus conventional pancreatic ductal adenocarcinoma: A comprehensive review of clinical-pathological features, outcomes, and molecular insights." Int J Mol Sci 22 (2021): 6756.
  22. Google Scholar, Crossref Indexed at

  23. Furukawa, Toru. "Mechanisms of development and progression of pancreatic neoplasms." Pathol Int 72 (2022): 529-540.
  24. Google Scholar, Crossref Indexed at

  25. Hirono, Seiko and Hiroki Yamaue. "Surgical strategy for intraductal papillary mucinous neoplasms of the pancreas." Surg Today 50 (2020): 50-55.
  26. Google Scholar, Crossref Indexed at

  27. Salvia, Roberto, Anna Burelli, Giampaolo Perri and Giovanni Marchegiani. "State-of-the-art surgical treatment of IPMNs." Langenbeck's Arch Surg (2021): 1-10.
  28. Google Scholar, Crossref Indexed at

  29. Aronsson, Linus, Sofia Marinko, Daniel Ansari and Roland Andersson. "Adjuvant therapy in invasive intraductal papillary mucinous neoplasm (IPMN) of the pancreas: A systematic review." Ann Transl Med 7 (2019): 689.
  30. Google Scholar, Crossref Indexed at

  31. Chong, Eric, Bathiya Ratnayake, Bobby VM Dasari and Benjamin PT Loveday, et al. "Adjuvant chemotherapy in the treatment of intraductal papillary mucinous neoplasms of the pancreas: Systematic review and meta-analysis." World J Surg (2022): 1-12.
  32. Google Scholar, Crossref Indexed at

  33. Kaiser, Joerg, Cornelius Scheifele, Ulf Hinz and Carl-Stephan Leonhardt, et al. "IPMN-associated pancreatic cancer: Survival, prognostic staging and impact of adjuvant chemotherapy." Eur J Surg Oncol 48 (2022): 1309-1320.
  34. Google Scholar, Crossref, Indexed at

  35. Sakitani, Mitsuru. "Multifaceted roles of carbon anhydrase IX in cancer cell proliferation, survival, metastasis and therapy resistance and indication of promising novel therapies by its inhibitors." J Mol Genet Med 16 (2022): 582.
  36. Crossref

  37. Sakitani, Mitsuru and Junichiro Taki. "Subtypes of intraductal papillary mucinous neo-plasms of the pancreas and its prognosis with reference to early palliative care integrated with oncology." Vox Propria 14 (2015): 1-34.
  38. Amato, Eliana, Marco dal Molin, Andrea Mafficini and Jun Yu, et al. "Targeted next‐generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas." J Pathol 233 (2014): 217-227.
  39. Google Scholar, Crossref Indexed at

  40. Paini, Marina, Stefano Crippa, Stefano Partelli and Filippo Scopelliti, et al. "Molecular pathology of intraductal papillary mucinous neoplasms of the pancreas." World J Gastroenterol 20 (2014): 10008.
  41. Google Scholar, Crossref Indexed at

  42. Pavlidis, Efstathios T., Konstantinos G. Sapalidis and Theodoros E. Pavlidis. "Modern aspects of the management of pancreatic intraductal papillary mucinous neoplasms: A narrative review." Rom J Morphol Embryol 63 (2022): 491-502.
  43. Google Scholar, Crossref Indexed at

  44. Turner, Ronald C., Jared T. Melnychuk, Wei Chen and Daniel Jones, et al. "Molecular analysis of pancreatic cyst Fluid for the management of intraductal papillary mucinous neoplasms." Diagnostics 12 (2022): 2573.
  45. Google Scholar, Crossref Indexed at

  46. Schönleben, Frank, Wanglong Qiu, Karl C. Bruckman and Nancy T. Ciau, et al. "BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas." Cancer Lett 249 (2007): 242-248.
  47. Google Scholar, Crossref, Indexed at

  48. Schönleben, Frank, Wanglong Qiu, Helen E. Remotti and Werner Hohenberger, et al. "PIK3CA, KRAS, and BRAF mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/C) of the pancreas." Langenbecks Arch Surg 393 (2008): 289-296.
  49. Google Scholar, Crossref, Indexed at

  50. Furukawa, Toru, Yuko Kuboki, Etsuko Tanji and Shoko Yoshida, et al. "Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas." Sci Rep 1 (2011): 161.
  51. Google Scholar, Crossref, Indexed at

  52. Matthaei, Hanno, Jian Wu, Marco Dal Molin and Chanjuan Shi, et al. "GNAS sequencing identifies IPMN-specific mutations in a subgroup of diminutive pancreatic cysts referred to as “incipient IPMNs”. Am J Surg Pathol 38 (2014): 360.
  53. Google Scholar, Crossref, Indexed at

  54. Tan, Marcus C., Olca Basturk, A. Rose Brannon and Umesh Bhanot, et al. "GNAS and KRAS mutations define separate progression pathways in intraductal papillary mucinous neoplasm-associated carcinoma." J Am Coll Surg 220 (2015): 845-854.
  55. Google Scholar, Crossref, Indexed at

  56. Chang, Xiao Yan, Yan Wu, Yuan Li and Jing Wang, et al. "Intraductal papillary mucinous neoplasms of the pancreas: Clinical association with KRAS." Mol Med Rep 17 (2018): 8061-8068.
  57. Google Scholar, Crossref, Indexed at

  58. Chang, Xiao Yan, Yan Wu, Ying Jiang and Peng Yan Wang, et al. "RNF43 mutations in IPMN cases: A potential prognostic factor." Gastroenterol Res Pract 2020 (2020):1457452.
  59. Google Scholar, Crossref, Indexed at

  60. McDonald, Paul C., Shawn C. Chafe, Wells S. Brown and Saeed Saberi, et al. "Regulation of pH by carbonic anhydrase 9 mediates survival of pancreatic cancer cells with activated KRAS in response to hypoxia." Gastroenterol 157 (2019): 823-837.
  61. Google Scholar, Crossref, Indexed at

  62. Furukawa, Toru, Makoto Sunamura, Fuyuhiko Motoi and Seiki Matsuno, et al. "Potential tumor suppressive pathway involving DUSP6/MKP-3 in pancreatic cancer." Am J Pathol 162 (2003): 1807-1815.
  63. Google Scholar, Crossref, Indexed at

  64. Lu, Zhimin and Shuichan Xu. "ERK1/2 MAP kinases in cell survival and apoptosis." IUBMB life 58 (2006): 621-631.
  65. Google Scholar, Crossref, Indexed at

  66. Furukawa, Toru. "Impacts of activation of the mitogen-activated protein kinase pathway in pancreatic cancer." Front Oncol 5 (2015): 23.
  67. Google Scholar, Crossref, Indexed at

  68. Jing, Yi, Ling-Zhi Liu, Yue Jiang and Yingxue Zhu, et al. "Cadmium increases HIF-1 and VEGF expression through ROS, ERK, and AKT signaling pathways and induces malignant transformation of human bronchial epithelial cells." Toxicol Sci 125 (2012): 10-19.
  69. Google Scholar, Crossref, Indexed at

  70. Masoud, Georgina N. and Wei Li. "HIF-1α pathway: Role, regulation and intervention for cancer therapy." Acta Pharm Sin B 5 (2015): 378-389.
  71. Google Scholar, Crossref, Indexed at

  72. Wan, Jun, and Wei Wu. "Hyperthermia induced HIF-1a expression of lung cancer through AKT and ERK signaling pathways." J Exp Clin Cancer Res 35 (2016): 1-11.
  73. Google Scholar, Crossref, Indexed at

  74. Xu, Xuewen, Kai You and Renge Bu. "Proximal tubular development is impaired with downregulation of MAPK/ERK signaling, HIF-1α, and catalase by hyperoxia exposure in neonatal rats." Oxid Med Cell Longev 2019 (2019): 9219847.
  75. Google Scholar, Crossref, Indexed at

  76. Kodaki, Tsutomu, Rüdiger Woscholski, Bengt Hallberg and Pablo Rodriguez-Viciana Julian Downward, et al. "The activation of phosphatidylinositol 3-kinase by Ras." Curr Biol 4 (1994): 798-806.
  77. Google Scholar, Crossref, Indexed at

  78. Rodriguez‐Viciana, P., P. H. Warne, B. Vanhaesebroeck and M. D. Waterfield, et al. "Activation of phosphoinositide 3‐kinase by interaction with Ras and by point mutation." EMBO J 15 (1996): 2442-2451.
  79. Google Scholar, Crossref, Indexed at

  80. Martinez, Nicholas G., David F. Thieker, Leiah M. Carey and Juhi A. Rasquinha, et al. "Biophysical and structural characterization of novel RAS-binding domains (RBDs) of PI3Kα and PI3Kγ." J Mol Biol 433 (2021): 166838.
  81. Google Scholar, Crossref, Indexed at

  82. O'hayre, Morgan, José Vázquez-Prado, Irina Kufareva and Eric W. Stawiski, et al. "The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer." Nat Rev Cancer 13 (2013): 412-424.
  83. Google Scholar, Crossref, Indexed at

  84. Patra, Krushna C., Yasutaka Kato, Yusuke Mizukami and Sebastian Widholz, et al. "Mutant GNAS drives pancreatic tumourigenesis by inducing PKA-mediated SIK suppression and reprogramming lipid metabolism." Nat Cell Biol 20 (2018): 811-822.
  85. Google Scholar, Crossref, Indexed at

  86. Chaudhary, Preeti Kumari and Soochong Kim. "An insight into GPCR and G-proteins as cancer drivers." Cells 10 (2021): 3288.
  87. Google Scholar, Crossref, Indexed at

  88. Zhang, Hongying, Qingbin Kong, Jiao Wang and Yangfu Jiang, et al. "Complex roles of cAMP–PKA–CREB signaling in cancer." Exp Hematol Oncol 9 (2020): 1-13.
  89. Google Scholar, Crossref, Indexed at

  90. Patra, Krushna C., Nabeel Bardeesy and Yusuke Mizukami. "Diversity of precursor lesions for pancreatic cancer: the genetics and biology of intraductal papillary mucinous neoplasm." Clin Transl Gastroenterol 8 (2017): e86.
  91. Google Scholar, Crossref, Indexed at

  92. Kuboki, Yuko, Catherine G. Fischer, Violeta Beleva Guthrie and Wenjie Huang, et al. "Single‐cell sequencing defines genetic heterogeneity in pancreatic cancer precursor lesions." J Pathol 247 (2019): 347-356.
  93. Google Scholar, Crossref, Indexed at

  94. Fischer, Catherine G., Violeta Beleva Guthrie, Alicia M. Braxton and Lily Zheng, et al. "Intraductal papillary mucinous neoplasms arise from multiple independent clones, each with distinct mutations." Gastroenterol 157 (2019): 1123-1137.
  95. Google Scholar, Crossref, Indexed at

  96. Rodriguez-Viciana, Pablo, Patricia H. Warne, Ritu Dhand and Bart Vanhaesebroeck, et al. "Phosphatidylinositol-3-OH kinase direct target of Ras." Nature 370 (1994): 527-532.
  97. Google Scholar, Crossref, Indexed at

  98. McIlroy, James, Daxin Chen, Christina Wjasow and Tamar Michaeli, et al. "Specific activation of p85-p110 phosphatidylinositol 3'-kinase stimulates DNA synthesis by ras-and p70 S6 kinase-dependent pathways." Mol Cell Biol 17 (1997): 248-255.
  99. Google Scholar, Crossref, Indexed at

  100. Huang, Chuan-Hsiang, Diana Mandelker, Oleg Schmidt-Kittler and Yardena Samuels, et al. "The structure of a human p110α/p85α complex elucidates the effects of oncogenic PI3Kα mutations." Science 318 (2007): 1744-1748.
  101. Google Scholar, Crossref, Indexed at

  102. Ali, Muhammad Umar, Muhammad Saif Ur Rahman, Zhenyu Jia and Cao Jiang. "Eukaryotic translation initiation factors and cancer." Tumour Biol 39 (2017): 1010428317709805.
  103. Google Scholar, Crossref, Indexed at

  104. Roux, Philippe P. and Ivan Topisirovic. "Signaling pathways involved in the regulation of mRNA translation." Mol Cell Biol 38 (2018): e00070-18.
  105. Google Scholar, Crossref, Indexed at

  106. Hao, Peiqi, Jiaojiao Yu, Richard Ward and Yin Liu, et al. "Eukaryotic translation initiation factors as promising targets in cancer therapy." Cell Commun Signal 18 (2020): 1-20.
  107. Google Scholar, Crossref, Indexed at

  108. Sang, Nianli, Daniel P. Stiehl, Jolene Bohensky and Irene Leshchinsky, et al. "MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300." J Biol Chem 278 (2003): 14013-14019.
  109. Google Scholar, Crossref, Indexed at

  110. Wykoff, Charles C., Nigel JP Beasley, Peter H. Watson and Kevin J. Turner, et al. "Hypoxia-inducible expression of tumor-associated carbonic anhydrases." Cancer res 60 (2000): 7075-7083.
  111. Google Scholar, Crossref, Indexed at

  112. Kaluz, Stefan, Milota Kaluzová and Eric J. Stanbridge. "Expression of the hypoxia marker carbonic anhydrase IX is critically dependent on SP1 activity. Identification of a novel type of hypoxia-responsive enhancer." Cancer res 63 (2003): 917-922.
  113. Google Scholar, Indexed at

  114. Kaluz, Stefan, Milota Kaluzová and Eric J. Stanbridge. "Regulation of gene expression by hypoxia: Integration of the HIF-transduced hypoxic signal at the hypoxia-responsive element." Clinica Chimica Acta 395 (2008): 6-13.
  115. Google Scholar, Crossref, Indexed at

  116. Chiche, Johanna, Karine Ilc, Julie Laferriere and Eric Trottier, et al. "Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH." Cancer res 69 (2009): 358-368.
  117. Google Scholar, Crossref, Indexed at

  118. Swietach, Pawel, Shalini Patiar, Claudiu T. Supuran and Adrian L. Harris, et al. "The role of carbonic anhydrase 9 in regulating extracellular and intracellular ph in three-dimensional tumor cell growths." J Biol Chem 284 (2009): 20299-20310.
  119. Google Scholar, Crossref, Indexed at

  120. Mohammad, Ramzi M., Irfana Muqbil, Leroy Lowe and Clement Yedjou, et al. "Broad targeting of resistance to apoptosis in cancer." Semin Cancer Biol 35 (2015): S78-S103.
  121. Google Scholar, Crossref, Indexed at

  122. Morana, Ornella, Will Wood and Christopher D. Gregory. "The apoptosis paradox in cancer." Int J Mol Sci 23 (2022): 1328.
  123. Google Scholar, Crossref, Indexed at

  124. Csaderova, Lucia, Michaela Debreova, Peter Radvak and Matej Stano, et al. "The effect of carbonic anhydrase IX on focal contacts during cell spreading and migration." Front Physiol 4 (2013): 271.
  125. Google Scholar, Crossref, Indexed at

  126. Mboge, Mam Y., Anusha Kota, Robert McKenna and Susan C. Frost. "Biophysical, biochemical, and cell based approaches used to decipher the role of carbonic anhydrases in cancer and to evaluate the potency of targeted inhibitors." Int J Med Chem 2018 (2018).
  127. Google Scholar, Crossref, Indexed at

  128. McDonald, Paul C., Mridula Swayampakula and Shoukat Dedhar. "Coordinated regulation of metabolic transporters and migration/invasion by carbonic anhydrase IX." Metabolites 8 (2018): 20.
  129. Google Scholar, Crossref, Indexed at

  130. Wouters, An, Bea Pauwels, Filip Lardon and Jan B. Vermorken. "Implications of in vitro research on the effect of radiotherapy and chemotherapy under hypoxic conditions." Oncologist 12 (2007): 690-712.
  131. Google Scholar, Crossref, Indexed at

  132. Tan, E. Y., M. Yan, L. Campo and C. Han, et al. "The key hypoxia regulated gene CAIX is upregulated in basal-like breast tumours and is associated with resistance to chemotherapy." Br J Cancer 100 (2009): 405-411.
  133. Google Scholar, Crossref, Indexed at

  134. Lock, F. E., P. C. McDonald, Y. Lou and I. Serrano, et al. "Targeting carbonic anhydrase IX depletes breast cancer stem cells within the hypoxic niche." Oncogene 32 (2013): 5210-5219.
  135. Google Scholar, Crossref, Indexed at

  136. Nasu, Kentaro, Kazunori Yamaguchi, Tomoka Takanashi and Keiichi Tamai, et al. "Crucial role of carbonic anhydrase IX in tumorigenicity of xenotransplanted adult T‐cell leukemia‐derived cells." Cancer Sci 108 (2017): 435-443.
  137. Google Scholar, Crossref, Indexed at

  138. Zeitouni, Daniel, Yuliya Pylayeva-Gupta, Channing J. Der and Kirsten L. Bryant. "KRAS mutant pancreatic cancer: No lone path to an effective treatment." Cancers 8 (2016): 45.
  139. Google Scholar, Crossref, Indexed at

  140. Waters, Andrew M and Channing J. Der. "KRAS: The critical driver and therapeutic target for pancreatic cancer." Cold    Spring Harb 8 (2018): a031435
  141. Google Scholar, Crossref, Indexed at

  142. Logsdon, Derek P., Michelle Grimard, Meihua Luo and Safi Shahda, et al. "Regulation of HIF1α under Hypoxia by APE1/Ref-1 Impacts CA9 Expression: Dual targeting in patient-derived 3d pancreatic cancer models dual-targeting ape1/ref-1 and ca9 in hypoxic pdac cells." Mol Cancer Ther 15 (2016): 2722-2732.
  143. Google Scholar, Crossref, Indexed at

  144. Logsdon, Derek P., Fenil Shah, Fabrizio Carta and Claudiu T. Supuran, et al. "Blocking HIF signaling via novel inhibitors of CA9 and APE1/Ref-1 dramatically affects pancreatic cancer cell survival." Sci Rep 8 (2018): 1-14.
  145. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3919

Molecular and Genetic Medicine received 3919 citations as per Google Scholar report

Molecular and Genetic Medicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward