Skip to main content

Pancreatic Diseases: The Role of Stem Cells

  • Chapter
  • First Online:
Digestive System Diseases

Part of the book series: Stem Cell Biology and Regenerative Medicine ((STEMCELL))

  • 478 Accesses

Abstract

Mesenchymal stem cells (MSCs) may be isolated from adipose tissue, bone marrow, umbilical cord, and peripheral blood. They are characterized by their innate ability to self-renew as well as differentiate into a variety of cell types. Several studies have demonstrated the innate characteristic of MSCs to home to sites of injury, inflammation, ischemia, as well as to tumors and metastases, including pancreatic gland inflammatory processes and pancreatic carcinoma, which renders them as an attractive option for cell-based therapy of these conditions.

Mesenchymal stem cells may have a crucial role in cell-based therapy of tumors, by specifically targeting certain aspects of tumor’s biology, either by introducing into the tumor genes, which interfere with specific molecular pathways or induce apoptosis, or by introducing specific genes, which contribute to the local activation of systemically administered prodrugs, avoiding thus the side effects, which are caused by the systemic administration of chemotherapy.

Recent studies have demonstrated that the inhibition of the anti-apoptotic X-linked inhibitor of apoptosis protein (XIAP), by using RNA interference, causes an enhanced TRAIL (tumor necrosis factor-related apoptosis-inducing ligand)-induced apoptosis in pancreatic cancer cells, both in vitro and in vivo. Moreover, by combining XIAP inhibition and human MSCs (hMSCs) expressing sTRAIL (the soluble form of TRAIL), the apoptotic activity was not only limited to slowing the tumor growth but also caused tumor’s remission and inhibition of metastatic growth. Alternatively, the production of IFN-b within the tumor site, by genetically modified MSCs, has been shown to suppress tumor growth to a significant extent.

The concept of gene-directed enzyme prodrug therapy, also called suicide gene therapy, in cancer therapy, has been under investigation in recent years. This approach consists of two steps: firstly, the use of genetically engineered MSCs as carriers of a specific gene that encodes a prodrug-activating protein and, secondly, the systematic administration of that particular prodrug that will be metabolized and activated into cytotoxic metabolites within the tumor. Given that angiogenesis is essential for tumor initiation and growth, genetically modified MSCs that express a therapeutic transgene under the control of TIE2 promoter upon reaching the tumor and in the presence of angiopoietin – 2 (ANGPT2) ligand, have been implicated in targeting the angiogenesis pathway, with the herpes simplex virus thymidine kinase (HSV-tk) being the most commonly used transgene. This mechanism of action, combined with the prodrug ganciclovir (GCV), resulted in a significant decrease in the volume of the pancreatic tumor. Alternatively, CC chemokine ligand 5 (CCL5) represents, among other chemokines, an essential factor in the homing process of MSCs to sites of tissue injury, including tumors and their metastases. Genetically modified MSCs to express the HSV-tk transgene under the control of the CCL5 promoter, combined with GCV as a prodrug, resulted in a significant decrease in the growth of the primary pancreatic tumor, as well as in a significant decrease in the incidence of metastatic lesions, in a mouse orthotopic model of pancreatic carcinoma.

In the context of acute pancreatitis, bone marrow MSCs (BM-MSCs) were the most commonly used MSCs, with nearly all studies demonstrating the immunomodulatory effect of BM-MSCs, by downregulating the expression of several pro-inflammatory markers and cytokines. Apart from their immunomodulatory effects, another potential mechanism of action of BM-MSCs is their antioxidant activities, by increasing the expression of glutathione peroxidase and superoxide dismutase. The therapeutic effect of BM-MSCs in acute pancreatitis is not only limited to the pancreatic gland, as BM-MSCs exert also their effects in other organs, including the improvement in the small intestinal capillary endothelial barrier.

On the other hand, only a limited number of studies have investigated the potential effect of MSCs administration for the treatment of chronic pancreatitis. Apart from the immunomodulatory effects of MSCs, all studies demonstrated a decrease both in the pancreatic damage and in the pancreatic fibrosis, following the administration of MSCs. Moreover, the administered BM-MSCs and umbilical cord-derived MSCs exerted an anti-apoptotic effect, by decreasing the apoptotic rate of the pancreatic acinar cells, as has been shown in two studies.

To date, there are certain limitations of cell-based therapies, which need to be carefully considered and require further investigation. First but not least, stem cells for therapeutic application need to meet the standards of Good Manufacturing Practice regulations, posing as important quality criteria, among others, the immunophenotype of the cells, the composition of the culture medium, and the risk for malignant transformation, as well as the aging and the immunosuppressive potential of the manufactured MSCs. Another issue regards the correct amount of MSCs that should be systemically administered, so as to home to the targeted tumor and exert their therapeutic effect. Several studies have focused on that issue, with some studies estimating the necessary amount of administered MSCs to be less than 10% of the targeted tumor mass.

At this point, it should be emphasized that the majority of studies investigating the insertional mutagenesis phenomenon are performed in rodent models, which have a relatively short life span, so that the true mutagenic risk cannot be estimated in the long-term. Therefore, the use of primate animal models, with a relatively longer life span, which may be administered greater amounts of MSCs, and possibly in a repeated fashion, will elucidate the true mutagenic risk of MSCs used for cell-based therapies.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 99.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 179.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.

    Article  CAS  PubMed  Google Scholar 

  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7–30.

    Article  PubMed  Google Scholar 

  3. Bosetti C, Bertuccio P, Malvezzi M, Levi F, Chatenoud L, Negri E, et al. Cancer mortality in Europe, 2005–2009, and an overview of trends since 1980. Ann Oncol. 2013;24(10):2657–71.

    Article  CAS  PubMed  Google Scholar 

  4. Bosetti C, Bertuccio P, Negri E, La Vecchia C, Zeegers MP, Boffetta P. Pancreatic cancer: overview of descriptive epidemiology. Mol Carcinog. 2012;51(1):3–13.

    Article  CAS  PubMed  Google Scholar 

  5. Herman JM, Swartz MJ, Hsu CC, Winter J, Pawlik TM, Sugar E, et al. Analysis of fluorouracil-based adjuvant chemotherapy and radiation after pancreaticoduodenectomy for ductal adenocarcinoma of the pancreas: results of a large, prospectively collected database at the Johns Hopkins Hospital. J Clin Oncol. 2008;26(21):3503–10.

    Article  PubMed  Google Scholar 

  6. Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, et al. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 2011;377(9765):557–67.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.

    Article  CAS  PubMed  Google Scholar 

  8. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.

    Article  CAS  Google Scholar 

  9. Loehrer PJ Sr, Feng Y, Cardenes H, Wagner L, Brell JM, Cella D, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an Eastern Cooperative Oncology Group trial. J Clin Oncol. 2011;29(31):4105–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Moorcraft SY, Khan K, Peckitt C, Watkins D, Rao S, Cunningham D, et al. FOLFIRINOX for locally advanced or metastatic pancreatic ductal adenocarcinoma: the Royal Marsden experience. Clin Colorectal Cancer. 2014;13(4):232–8.

    Article  PubMed  Google Scholar 

  11. Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350(12):1200–10.

    Article  CAS  PubMed  Google Scholar 

  12. Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304(10):1073–81.

    Article  CAS  PubMed  Google Scholar 

  13. Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297(3):267–77.

    Article  CAS  PubMed  Google Scholar 

  14. Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310(14):1473–81.

    Article  CAS  PubMed  Google Scholar 

  15. Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18(5):1319–26.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Bruce WR, Van Der Gaag H. A quantitative assay for the number of murine lymphoma cells capable of proliferation in vivo. Nature. 1963;199:79–80.

    Article  CAS  PubMed  Google Scholar 

  17. Park CH, Bergsagel DE, McCulloch EA. Mouse myeloma tumor stem cells: a primary cell culture assay. J Natl Cancer Inst. 1971;46(2):411–22.

    CAS  PubMed  Google Scholar 

  18. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67(3):1030–7.

    Article  CAS  PubMed  Google Scholar 

  19. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401.

    Article  CAS  PubMed  Google Scholar 

  21. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445(7123):111–5.

    Article  CAS  PubMed  Google Scholar 

  22. Jordan CT. Cancer stem cells: controversial or just misunderstood? Cell Stem Cell. 2009;4(3):203–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1(3):313–23.

    Article  CAS  PubMed  Google Scholar 

  24. Li X, Zhao H, Gu J, Zheng L. Prognostic value of cancer stem cell marker CD133 expression in pancreatic ductal adenocarcinoma (PDAC): a systematic review and meta-analysis. Int J Clin Exp Pathol. 2015;8(10):12084–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Maeda S, Shinchi H, Kurahara H, Mataki Y, Maemura K, Sato M, et al. CD133 expression is correlated with lymph node metastasis and vascular endothelial growth factor-C expression in pancreatic cancer. Br J Cancer. 2008;98(8):1389–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li C, Wu JJ, Hynes M, Dosch J, Sarkar B, Welling TH, et al. c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology. 2011;141(6):2218–2227 e2215.

    Article  CAS  PubMed  Google Scholar 

  27. Challen GA, Little MH. A side order of stem cells: the SP phenotype. Stem Cells. 2006;24(1):3–12.

    Article  PubMed  Google Scholar 

  28. Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, et al. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci U S A. 2004;101(39):14228–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 1996;183(4):1797–806.

    Article  CAS  PubMed  Google Scholar 

  30. Chuthapisith S, Eremin JM, Eremin O. Predicting response to neoadjuvant chemotherapy in breast cancer: molecular imaging, systemic biomarkers and the cancer metabolome (review). Oncol Rep. 2008;20(4):699–703.

    PubMed  Google Scholar 

  31. Steiniger SC, Coppinger JA, Kruger JA, Yates J 3rd, Janda KD. Quantitative mass spectrometry identifies drug targets in cancer stem cell-containing side population. Stem Cells. 2008;26(12):3037–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hong SP, Wen J, Bang S, Park S, Song SY. CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer. 2009;125(10):2323–31.

    Article  CAS  PubMed  Google Scholar 

  33. Rasheed ZA, Yang J, Wang Q, Kowalski J, Freed I, Murter C, et al. Prognostic significance of tumorigenic cells with mesenchymal features in pancreatic adenocarcinoma. J Natl Cancer Inst. 2010;102(5):340–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Green MR. Targeting targeted therapy. N Engl J Med. 2004;350(21):2191–3.

    Article  CAS  PubMed  Google Scholar 

  35. Corsten MF, Shah K. Therapeutic stem-cells for cancer treatment: hopes and hurdles in tactical warfare. Lancet Oncol. 2008;9(4):376–84.

    Article  PubMed  Google Scholar 

  36. Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev. 2012;64(8):739–48.

    Article  CAS  PubMed  Google Scholar 

  37. Yip S, Shah K. Stem-cell based therapies for brain tumors. Curr Opin Mol Ther. 2008;10(4):334–42.

    PubMed  Google Scholar 

  38. Sagar J, Chaib B, Sales K, Winslet M, Seifalian A. Role of stem cells in cancer therapy and cancer stem cells: a review. Cancer Cell Int. 2007;7:9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Spring H, Schuler T, Arnold B, Hammerling GJ, Ganss R. Chemokines direct endothelial progenitors into tumor neovessels. Proc Natl Acad Sci U S A. 2005;102(50):18111–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cihova M, Altanerova V, Altaner C. Stem cell based cancer gene therapy. Mol Pharm. 2011;8(5):1480–7.

    Article  CAS  PubMed  Google Scholar 

  41. Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, Meyn RE, et al. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res. 2007;67(24):11687–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ, Andreeff M. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res. 2002;62(13):3603–8.

    CAS  PubMed  Google Scholar 

  43. Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-Hansen M, Bekele BN, et al. Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst. 2004;96(21):1593–603.

    Article  CAS  PubMed  Google Scholar 

  44. Dwyer RM, Khan S, Barry FP, O’Brien T, Kerin MJ. Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Res Ther. 2010;1(3):25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum Gene Ther. 2010;21(11):1513–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim SU, Jeung EB, Kim YB, Cho MH, Choi KC. Potential tumor-tropic effect of genetically engineered stem cells expressing suicide enzymes to selectively target invasive cancer in animal models. Anticancer Res. 2011;31(4):1249–58.

    PubMed  Google Scholar 

  47. Kucerova L, Altanerova V, Matuskova M, Tyciakova S, Altaner C. Adipose tissue-derived human mesenchymal stem cells mediated prodrug cancer gene therapy. Cancer Res. 2007;67(13):6304–13.

    Article  CAS  PubMed  Google Scholar 

  48. Morizono K, De Ugarte DA, Zhu M, Zuk P, Elbarbary A, Ashjian P, et al. Multilineage cells from adipose tissue as gene delivery vehicles. Hum Gene Ther. 2003;14(1):59–66.

    Article  CAS  PubMed  Google Scholar 

  49. Meyerrose TE, De Ugarte DA, Hofling AA, Herrbrich PE, Cordonnier TD, Shultz LD, et al. In vivo distribution of human adipose-derived mesenchymal stem cells in novel xenotransplantation models. Stem Cells. 2007;25(1):220–7.

    Article  CAS  PubMed  Google Scholar 

  50. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25(11):2739–49.

    Article  CAS  PubMed  Google Scholar 

  51. Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cells derived from various mesenchymal tissues: superiority of synovium as a cell source. Arthritis Rheum. 2005;52(8):2521–9.

    Article  PubMed  Google Scholar 

  52. Altaner C. Prodrug cancer gene therapy. Cancer Lett. 2008;270(2):191–201.

    Article  CAS  PubMed  Google Scholar 

  53. Moniri MR, Sun XY, Rayat J, Dai D, Ao Z, He Z, et al. TRAIL-engineered pancreas-derived mesenchymal stem cells: characterization and cytotoxic effects on pancreatic cancer cells. Cancer Gene Ther. 2012;19(9):652–8.

    Article  CAS  PubMed  Google Scholar 

  54. Zischek C, Niess H, Ischenko I, Conrad C, Huss R, Jauch KW, et al. Targeting tumor stroma using engineered mesenchymal stem cells reduces the growth of pancreatic carcinoma. Ann Surg. 2009;250(5):747–53.

    Article  PubMed  Google Scholar 

  55. Menon LG, Picinich S, Koneru R, Gao H, Lin SY, Koneru M, et al. Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells. Stem Cells. 2007;25(2):520–8.

    Article  CAS  PubMed  Google Scholar 

  56. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell. 2009;4(3):206–16.

    Article  CAS  PubMed  Google Scholar 

  57. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med. 2001;7(11):1194–201.

    Article  CAS  PubMed  Google Scholar 

  58. Beckermann BM, Kallifatidis G, Groth A, Frommhold D, Apel A, Mattern J, et al. VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer. 2008;99(4):622–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sasportas LS, Kasmieh R, Wakimoto H, Hingtgen S, van de Water JA, Mohapatra G, et al. Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy. Proc Natl Acad Sci U S A. 2009;106(12):4822–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, et al. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci U S A. 2004;101(52):18117–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schmidt NO, Przylecki W, Yang W, Ziu M, Teng Y, Kim SU, et al. Brain tumor tropism of transplanted human neural stem cells is induced by vascular endothelial growth factor. Neoplasia. 2005;7(6):623–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Son BR, Marquez-Curtis LA, Kucia M, Wysoczynski M, Turner AR, Ratajczak J, et al. Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases. Stem Cells. 2006;24(5):1254–64.

    Article  CAS  PubMed  Google Scholar 

  63. Andoh A, Takaya H, Saotome T, Shimada M, Hata K, Araki Y, et al. Cytokine regulation of chemokine (IL-8, MCP-1, and RANTES) gene expression in human pancreatic periacinar myofibroblasts. Gastroenterology. 2000;119(1):211–9.

    Article  CAS  PubMed  Google Scholar 

  64. Azenshtein E, Luboshits G, Shina S, Neumark E, Shahbazian D, Weil M, et al. The CC chemokine RANTES in breast carcinoma progression: regulation of expression and potential mechanisms of promalignant activity. Cancer Res. 2002;62(4):1093–102.

    CAS  PubMed  Google Scholar 

  65. Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008;267(2):271–85.

    Article  CAS  PubMed  Google Scholar 

  66. Ruster B, Gottig S, Ludwig RJ, Bistrian R, Muller S, Seifried E, et al. Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood. 2006;108(12):3938–44.

    Article  PubMed  CAS  Google Scholar 

  67. Jin H, Aiyer A, Su J, Borgstrom P, Stupack D, Friedlander M, et al. A homing mechanism for bone marrow-derived progenitor cell recruitment to the neovasculature. J Clin Invest. 2006;116(3):652–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Motaln H, Schichor C, Lah TT. Human mesenchymal stem cells and their use in cell-based therapies. Cancer. 2010;116(11):2519–30.

    Article  CAS  PubMed  Google Scholar 

  69. Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65(8):3307–18.

    Article  CAS  PubMed  Google Scholar 

  70. Mohr A, Albarenque SM, Deedigan L, Yu R, Reidy M, Fulda S, et al. Targeting of XIAP combined with systemic mesenchymal stem cell-mediated delivery of sTRAIL ligand inhibits metastatic growth of pancreatic carcinoma cells. Stem Cells. 2010;28(11):2109–20.

    Article  CAS  PubMed  Google Scholar 

  71. Vogler M, Durr K, Jovanovic M, Debatin KM, Fulda S. Regulation of TRAIL-induced apoptosis by XIAP in pancreatic carcinoma cells. Oncogene. 2007;26(2):248–57.

    Article  CAS  PubMed  Google Scholar 

  72. Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, et al. Targeting XIAP bypasses Bcl-2-mediated resistance to TRAIL and cooperates with TRAIL to suppress pancreatic cancer growth in vitro and in vivo. Cancer Res. 2008;68(19):7956–65.

    Article  CAS  PubMed  Google Scholar 

  73. Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, et al. Small molecule XIAP inhibitors enhance TRAIL-induced apoptosis and antitumor activity in preclinical models of pancreatic carcinoma. Cancer Res. 2009;69(6):2425–34.

    Article  CAS  PubMed  Google Scholar 

  74. Lu YR, Yuan Y, Wang XJ, Wei LL, Chen YN, Cong C, et al. The growth inhibitory effect of mesenchymal stem cells on tumor cells in vitro and in vivo. Cancer Biol Ther. 2008;7(2):245–51.

    Article  CAS  PubMed  Google Scholar 

  75. Sun B, Roh KH, Park JR, Lee SR, Park SB, Jung JW, et al. Therapeutic potential of mesenchymal stromal cells in a mouse breast cancer metastasis model. Cytotherapy. 2009;11(3):289–98.. 281 p following 298

    Article  CAS  PubMed  Google Scholar 

  76. Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, et al. Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy. 2010;12(5):615–25.

    Article  CAS  PubMed  Google Scholar 

  77. Greco O, Dachs GU. Gene directed enzyme/prodrug therapy of cancer: historical appraisal and future prospectives. J Cell Physiol. 2001;187(1):22–36.

    Article  CAS  PubMed  Google Scholar 

  78. Bi WL, Parysek LM, Warnick R, Stambrook PJ. In vitro evidence that metabolic cooperation is responsible for the bystander effect observed with HSV tk retroviral gene therapy. Hum Gene Ther. 1993;4(6):725–31.

    Article  CAS  PubMed  Google Scholar 

  79. Gagandeep S, Brew R, Green B, Christmas SE, Klatzmann D, Poston GJ, et al. Prodrug-activated gene therapy: involvement of an immunological component in the “bystander effect”. Cancer Gene Ther. 1996;3(2):83–8.

    CAS  PubMed  Google Scholar 

  80. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72.

    Article  CAS  PubMed  Google Scholar 

  81. Kuriyama S, Tsujinoue H, Yoshiji H. Immune response to suicide gene therapy. Methods Mol Med. 2004;90:353–69.

    CAS  PubMed  Google Scholar 

  82. Touati W, Tran T, Seguin J, Diry M, Flinois JP, Baillou C, et al. A suicide gene therapy combining the improvement of cyclophosphamide tumor cytotoxicity and the development of an anti-tumor immune response. Curr Gene Ther. 2014;14(3):236–46.

    Article  CAS  PubMed  Google Scholar 

  83. Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh KR, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci. 2003;10(2):228–41.

    Article  CAS  PubMed  Google Scholar 

  84. Guo X, Evans TR, Somanath S, Armesilla AL, Darling JL, Schatzlein A, et al. In vitro evaluation of cancer-specific NF-kappaB-CEA enhancer-promoter system for 5-fluorouracil prodrug gene therapy in colon cancer cell lines. Br J Cancer. 2007;97(6):745–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Nyati MK, Sreekumar A, Li S, Zhang M, Rynkiewicz SD, Chinnaiyan AM, et al. High and selective expression of yeast cytosine deaminase under a carcinoembryonic antigen promoter-enhancer. Cancer Res. 2002;62(8):2337–42.

    CAS  PubMed  Google Scholar 

  86. Conrad C, Husemann Y, Niess H, von Luettichau I, Huss R, Bauer C, et al. Linking transgene expression of engineered mesenchymal stem cells and angiopoietin-1-induced differentiation to target cancer angiogenesis. Ann Surg. 2011;253(3):566–71.

    Article  PubMed  Google Scholar 

  87. Niess H, Bao Q, Conrad C, Zischek C, Notohamiprodjo M, Schwab F, et al. Selective targeting of genetically engineered mesenchymal stem cells to tumor stroma microenvironments using tissue-specific suicide gene expression suppresses growth of hepatocellular carcinoma. Ann Surg. 2011;254(5):767–74.. discussion 774–765

    Article  PubMed  Google Scholar 

  88. Huang H, Bhat A, Woodnutt G, Lappe R. Targeting the ANGPT-TIE2 pathway in malignancy. Nat Rev Cancer. 2010;10(8):575–85.

    Article  CAS  PubMed  Google Scholar 

  89. Herbst RS, Hong D, Chap L, Kurzrock R, Jackson E, Silverman JM, et al. Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. J Clin Oncol. 2009;27(21):3557–65.

    Article  CAS  PubMed  Google Scholar 

  90. Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology. 2013;144(6):1252–61.

    Article  PubMed  Google Scholar 

  91. Chan YC, Leung PS. Acute pancreatitis: animal models and recent advances in basic research. Pancreas. 2007;34(1):1–14.

    Article  PubMed  Google Scholar 

  92. Ishibashi T, Zhao H, Kawabe K, Oono T, Egashira K, Suzuki K, et al. Blocking of monocyte chemoattractant protein-1 (MCP-1) activity attenuates the severity of acute pancreatitis in rats. J Gastroenterol. 2008;43(1):79–85.

    Article  CAS  PubMed  Google Scholar 

  93. Frossard JL, Steer ML, Pastor CM. Acute pancreatitis. Lancet. 2008;371(9607):143–52.

    Article  PubMed  Google Scholar 

  94. Norman JG, Fink GW, Messina J, Carter G, Franz MG. Timing of tumor necrosis factor antagonism is critical in determining outcome in murine lethal acute pancreatitis. Surgery. 1996;120(3):515–21.

    Article  CAS  PubMed  Google Scholar 

  95. Huang LY, Chen P, Xu LX, Zhou YF, Zhang YP, Yuan YZ. Fractalkine upregulates inflammation through CX3CR1 and the Jak-Stat pathway in severe acute pancreatitis rat model. Inflammation. 2012;35(3):1023–30.

    Article  CAS  PubMed  Google Scholar 

  96. Huang L, Ma J, Tang Y, Chen P, Zhang S, Zhang Y, et al. siRNA-based targeting of fractalkine overexpression suppresses inflammation development in a severe acute pancreatitis rat model. Int J Mol Med. 2012;30(3):514–20.

    Article  CAS  PubMed  Google Scholar 

  97. Working Group IAPAPAAPG. IAP/APA evidence-based guidelines for the management of acute pancreatitis. Pancreatology. 2013;13(4 Suppl 2):e1–15.

    Google Scholar 

  98. Ranson JH, Rifkind KM, Roses DF, Fink SD, Eng K, Spencer FC. Prognostic signs and the role of operative management in acute pancreatitis. Surg Gynecol Obstet. 1974;139(1):69–81.

    CAS  PubMed  Google Scholar 

  99. Ranson JH, Pasternack BS. Statistical methods for quantifying the severity of clinical acute pancreatitis. J Surg Res. 1977;22(2):79–91.

    Article  CAS  PubMed  Google Scholar 

  100. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818–29.

    Article  CAS  PubMed  Google Scholar 

  101. Wilson C, Heath DI, Imrie CW. Prediction of outcome in acute pancreatitis: a comparative study of APACHE II, clinical assessment and multiple factor scoring systems. Br J Surg. 1990;77(11):1260–4.

    Article  CAS  PubMed  Google Scholar 

  102. Halonen KI, Pettila V, Leppaniemi AK, Kemppainen EA, Puolakkainen PA, Haapiainen RK. Multiple organ dysfunction associated with severe acute pancreatitis. Crit Care Med. 2002;30(6):1274–9.

    Article  PubMed  Google Scholar 

  103. Vincent JL, de Mendonca A, Cantraine F, Moreno R, Takala J, Suter PM, et al. Use of the SOFA score to assess the incidence of organ dysfunction/failure in intensive care units: results of a multicenter, prospective study. Working group on “sepsis-related problems” of the European Society of Intensive Care Medicine. Crit Care Med. 1998;26(11):1793–800.

    Article  CAS  PubMed  Google Scholar 

  104. Balthazar EJ, Robinson DL, Megibow AJ, Ranson JH. Acute pancreatitis: value of CT in establishing prognosis. Radiology. 1990;174(2):331–6.

    Article  CAS  PubMed  Google Scholar 

  105. Eachempati SR, Hydo LJ, Barie PS. Severity scoring for prognostication in patients with severe acute pancreatitis: comparative analysis of the Ranson score and the APACHE III score. Arch Surg. 2002;137(6):730–6.

    Article  PubMed  Google Scholar 

  106. Liu TH, Kwong KL, Tamm EP, Gill BS, Brown SD, Mercer DW. Acute pancreatitis in intensive care unit patients: value of clinical and radiologic prognosticators at predicting clinical course and outcome. Crit Care Med. 2003;31(4):1026–30.

    Article  PubMed  Google Scholar 

  107. Sheth SG, Conwell DL, Whitcomb DC, Alsante M, Anderson MA, Barkin J, et al. Academic pancreas centers of excellence: guidance from a multidisciplinary chronic pancreatitis working group at PancreasFest. Pancreatology. 2017;17(3):419–30.

    Article  PubMed  Google Scholar 

  108. Whitcomb DC, Shimosegawa T, Chari ST, Forsmark CE, Frulloni L, Garg P, et al. International consensus statements on early chronic Pancreatitis. Recommendations from the working group for the international consensus guidelines for chronic pancreatitis in collaboration with The International Association of Pancreatology, American Pancreatic Association, Japan Pancreas Society, PancreasFest Working Group and European Pancreatic Club. Pancreatology. 2018.

    Google Scholar 

  109. Linard C, Busson E, Holler V, Strup-Perrot C, Lacave-Lapalun JV, Lhomme B, et al. Repeated autologous bone marrow-derived mesenchymal stem cell injections improve radiation-induced proctitis in pigs. Stem Cells Transl Med. 2013;2(11):916–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Tsai PC, Fu TW, Chen YM, Ko TL, Chen TH, Shih YH, et al. The therapeutic potential of human umbilical mesenchymal stem cells from Wharton’s jelly in the treatment of rat liver fibrosis. Liver Transpl. 2009;15(5):484–95.

    Article  PubMed  Google Scholar 

  111. Song L, Yang YJ, Dong QT, Qian HY, Gao RL, Qiao SB, et al. Atorvastatin enhance efficacy of mesenchymal stem cells treatment for swine myocardial infarction via activation of nitric oxide synthase. PLoS One. 2013;8(5):e65702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Li L, Zhang Y, Li Y, Yu B, Xu Y, Zhao S, et al. Mesenchymal stem cell transplantation attenuates cardiac fibrosis associated with isoproterenol-induced global heart failure. Transpl Int. 2008;21(12):1181–9.

    Article  CAS  PubMed  Google Scholar 

  113. Jung KH, Song SU, Yi T, Jeon MS, Hong SW, Zheng HM, et al. Human bone marrow-derived clonal mesenchymal stem cells inhibit inflammation and reduce acute pancreatitis in rats. Gastroenterology. 2011;140(3):998–1008.

    Article  CAS  PubMed  Google Scholar 

  114. Jung KH, Yi T, Son MK, Song SU, Hong SS. Therapeutic effect of human clonal bone marrow-derived mesenchymal stem cells in severe acute pancreatitis. Arch Pharm Res. 2015;38(5):742–51.

    Article  CAS  PubMed  Google Scholar 

  115. Qian D, Gong J, He Z, Hua J, Lin S, Xu C, et al. Bone marrow-derived mesenchymal stem cells repair necrotic pancreatic tissue and promote angiogenesis by secreting cellular growth factors involved in the SDF-1 alpha/CXCR4 Axis in rats. Stem Cells Int. 2015;2015:306836.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. He Z, Hua J, Qian D, Gong J, Lin S, Xu C, et al. Intravenous hMSCs ameliorate acute pancreatitis in mice via secretion of tumor necrosis factor-alpha stimulated gene/protein 6. Sci Rep. 2016;6:38438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Yin G, Hu G, Wan R, Yu G, Cang X, Ni J, et al. Role of bone marrow mesenchymal stem cells in L-arg-induced acute pancreatitis: effects and possible mechanisms. Int J Clin Exp Pathol. 2015;8(5):4457–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhao H, He Z, Huang D, Gao J, Gong Y, Wu H, et al. Infusion of bone marrow mesenchymal stem cells attenuates experimental severe acute pancreatitis in rats. Stem Cells Int. 2016;2016:7174319.

    PubMed  PubMed Central  Google Scholar 

  119. Chen Z, Lu F, Fang H, Huang H. Effect of mesenchymal stem cells on renal injury in rats with severe acute pancreatitis. Exp Biol Med (Maywood). 2013;238(6):687–95.

    Article  CAS  Google Scholar 

  120. Tu XH, Song JX, Xue XJ, Guo XW, Ma YX, Chen ZY, et al. Role of bone marrow-derived mesenchymal stem cells in a rat model of severe acute pancreatitis. World J Gastroenterol. 2012;18(18):2270–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Qu B, Chu Y, Zhu F, Wang B, Liu T, Yu B, et al. Granulocyte colony-stimulating factor enhances the therapeutic efficacy of bone marrow mesenchymal stem cell transplantation in rats with experimental acute pancreatitis. Oncotarget. 2017;8(13):21305–14.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Qian D, Wei G, Xu C, He Z, Hua J, Li J, et al. Bone marrow-derived mesenchymal stem cells (BMSCs) repair acute necrotized pancreatitis by secreting microRNA-9 to target the NF-kappaB1/p50 gene in rats. Sci Rep. 2017;7(1):581.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Yin G, Hu G, Wan R, Yu G, Cang X, Xiong J, et al. Role of microvesicles from bone marrow mesenchymal stem cells in acute pancreatitis. Pancreas. 2016;45(9):1282–93.

    Article  PubMed  Google Scholar 

  124. Meng HB, Gong J, Zhou B, Hua J, Yao L, Song ZS. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells in rat severe acute pancreatitis. Int J Clin Exp Pathol. 2013;6(12):2703–12.

    PubMed  PubMed Central  Google Scholar 

  125. Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B, Hows JM. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121(2):368–74.

    Article  PubMed  Google Scholar 

  126. Lu F, Wang F, Chen Z, Huang H. Effect of mesenchymal stem cells on small intestinal injury in a rat model of acute necrotizing pancreatitis. Stem Cell Res Ther. 2017;8(1):12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Yang B, Bai B, Liu CX, Wang SQ, Jiang X, Zhu CL, et al. Effect of umbilical cord mesenchymal stem cells on treatment of severe acute pancreatitis in rats. Cytotherapy. 2013;15(2):154–62.

    Article  CAS  PubMed  Google Scholar 

  128. Hua J, He ZG, Qian DH, Lin SP, Gong J, Meng HB, et al. Angiopoietin-1 gene-modified human mesenchymal stem cells promote angiogenesis and reduce acute pancreatitis in rats. Int J Clin Exp Pathol. 2014;7(7):3580–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Kawakubo K, Ohnishi S, Fujita H, Kuwatani M, Onishi R, Masamune A, et al. Effect of fetal membrane-derived mesenchymal stem cell transplantation in rats with acute and chronic pancreatitis. Pancreas. 2016;45(5):707–13.

    Article  CAS  PubMed  Google Scholar 

  130. Kim HW, Song WJ, Li Q, Han SM, Jeon KO, Park SC, et al. Canine adipose tissue-derived mesenchymal stem cells ameliorate severe acute pancreatitis by regulating T cells in rats. J Vet Sci. 2016;17(4):539–48.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Chiesa S, Morbelli S, Morando S, Massollo M, Marini C, Bertoni A, et al. Mesenchymal stem cells impair in vivo T-cell priming by dendritic cells. Proc Natl Acad Sci U S A. 2011;108(42):17384–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Schrepfer S, Deuse T, Reichenspurner H, Fischbein MP, Robbins RC, Pelletier MP. Stem cell transplantation: the lung barrier. Transplant Proc. 2007;39(2):573–6.

    Article  CAS  PubMed  Google Scholar 

  133. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5(1):54–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Qin T, Liu CJ, Zhang HW, Pan YF, Tang Q, Liu JK, et al. Effect of the IkBalpha mutant gene delivery to mesenchymal stem cells on rat chronic pancreatitis. Genet Mol Res. 2014;13(1):371–85.

    Article  CAS  PubMed  Google Scholar 

  135. Zhou CH, Li ML, Qin AL, Lv SX, Wen T, Zhu XY, et al. Reduction of fibrosis in dibutyltin dichloride-induced chronic pancreatitis using rat umbilical mesenchymal stem cells from Wharton’s jelly. Pancreas. 2013;42(8):1291–302.

    Article  CAS  PubMed  Google Scholar 

  136. Wuchter P, Bieback K, Schrezenmeier H, Bornhauser M, Muller LP, Bonig H, et al. Standardization of good manufacturing practice-compliant production of bone marrow-derived human mesenchymal stromal cells for immunotherapeutic applications. Cytotherapy. 2015;17(2):128–39.

    Article  CAS  PubMed  Google Scholar 

  137. Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24(5):1294–301.

    Article  CAS  PubMed  Google Scholar 

  138. Zhang ZX, Guan LX, Zhang K, Zhang Q, Dai LJ. A combined procedure to deliver autologous mesenchymal stromal cells to patients with traumatic brain injury. Cytotherapy. 2008;10(2):134–9.

    Article  CAS  PubMed  Google Scholar 

  139. Li DS, Warnock GL, Tu HJ, Ao Z, He Z, Lu H, et al. Do immunotherapy and beta cell replacement play a synergistic role in the treatment of type 1 diabetes? Life Sci. 2009;85(15–16):549–56.

    Article  CAS  PubMed  Google Scholar 

  140. Hung SC, Deng WP, Yang WK, Liu RS, Lee CC, Su TC, et al. Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clin Cancer Res. 2005;11(21):7749–56.

    Article  CAS  PubMed  Google Scholar 

  141. Kim SW, Kim SJ, Park SH, Yang HG, Kang MC, Choi YW, et al. Complete regression of metastatic renal cell carcinoma by multiple injections of engineered mesenchymal stem cells expressing dodecameric TRAIL and HSV-TK. Clin Cancer Res. 2013;19(2):415–27.

    Article  CAS  PubMed  Google Scholar 

  142. Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, et al. Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood. 2003;102(10):3837–44.

    Article  CAS  PubMed  Google Scholar 

  143. Djouad F, Bony C, Apparailly F, Louis-Plence P, Jorgensen C, Noel D. Earlier onset of syngeneic tumors in the presence of mesenchymal stem cells. Transplantation. 2006;82(8):1060–6.

    Article  PubMed  Google Scholar 

  144. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.

    Article  CAS  PubMed  Google Scholar 

  145. Pike-Overzet K, van der Burg M, Wagemaker G, van Dongen JJ, Staal FJ. New insights and unresolved issues regarding insertional mutagenesis in X-linked SCID gene therapy. Mol Ther. 2007;15(11):1910–6.

    Article  CAS  PubMed  Google Scholar 

  146. Zhang TY, Huang B, Yuan ZY, Hu YL, Tabata Y, Gao JQ. Gene recombinant bone marrow mesenchymal stem cells as a tumor-targeted suicide gene delivery vehicle in pulmonary metastasis therapy using non-viral transfection. Nanomedicine. 2014;10(1):257–67.

    Article  CAS  PubMed  Google Scholar 

  147. Coffelt SB, Marini FC, Watson K, Zwezdaryk KJ, Dembinski JL, LaMarca HL, et al. The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proc Natl Acad Sci U S A. 2009;106(10):3806–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449(7162):557–63.

    Article  CAS  PubMed  Google Scholar 

  149. Zhu W, Xu W, Jiang R, Qian H, Chen M, Hu J, et al. Mesenchymal stem cells derived from bone marrow favor tumor cell growth in vivo. Exp Mol Pathol. 2006;80(3):267–74.

    Article  CAS  PubMed  Google Scholar 

  150. Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One. 2009;4(4):e4992.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Marofi, et al. Mesenchymal stromal/stem cells: a new era in the cell-based targeted gene therapy of cancer. Front Immunol. 2017;8:1770. https://doi.org/10.3389/fimmu.2017.01770.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Williams, et al. Nitroreductase gene-directed enzyme prodrug therapy: insights and advances toward clinical utility. Biochem J. 2015;471:131–53. https://doi.org/10.1042/BJ20150650.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Apostolou, K.G. (2019). Pancreatic Diseases: The Role of Stem Cells. In: Gazouli, M., Theodoropoulos, G. (eds) Digestive System Diseases. Stem Cell Biology and Regenerative Medicine. Humana Press, Cham. https://doi.org/10.1007/978-3-030-11965-2_4

Download citation

Publish with us

Policies and ethics