Summary

Reprogramming Pancreatic Ductal Adenocarcinoma to Pluripotency

Published: February 02, 2024
doi:

Summary

The present protocol describes the reprogramming of Pancreatic Ductal Adenocarcinoma (PDAC) and normal pancreatic ductal epithelial cells into induced pluripotent stem cells (iPSCs). We provide an optimized and detailed, step-by-step procedure, from preparing lentivirus to establishing stable iPSC lines.

Abstract

The generation of induced pluripotent stem cells (iPSCs) using transcription factors has been achieved from almost any differentiated cell type and has proved highly valuable for research and clinical applications. Interestingly, iPSC reprogramming of cancer cells, such as pancreatic ductal adenocarcinoma (PDAC), has been shown to revert the invasive PDAC phenotype and override the cancer epigenome. The differentiation of PDAC-derived iPSCs can recapitulate PDAC progression from its early pancreatic intraepithelial neoplasia (PanIN) precursor, revealing the molecular and cellular changes that occur early during PDAC progression. Therefore, PDAC-derived iPSCs can be used to model the earliest stages of PDAC for the discovery of early-detection diagnostic markers. This is particularly important for PDAC patients, who are typically diagnosed at the late metastatic stages due to a lack of reliable biomarkers for the earlier PanIN stages. However, reprogramming cancer cell lines, including PDAC, into pluripotency remains challenging, labor-intensive, and highly variable between different lines. Here, we describe a more consistent protocol for generating iPSCs from various human PDAC cell lines using bicistronic lentiviral vectors. The resulting iPSC lines are stable, showing no dependence on the exogenous expression of reprogramming factors or inducible drugs. Overall, this protocol facilitates the generation of a wide range of PDAC-derived iPSCs, which is essential for discovering early biomarkers that are more specific and representative of PDAC cases.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignancies, and early diagnosis remains challenging due to the asymptomatic nature of the disease. The majority of PDAC patients are diagnosed at the advanced metastatic stage when very limited treatment options are available1,2. This is mainly due to the lack of reliable biomarkers for the earlier stages, such as those that could be conveniently detected as proteins released into the bloodstream.

PDAC can disseminate very early during its progression, and a better prognosis has been linked to early cancer detection when PDAC is localized in the pancreas3. However, less than a tenth of PDAC patients are diagnosed with a favorable prognosis, allowing for surgical resection. Nonetheless, those few with resectable tumors are also prone to tumor recurrence within 12 months4.

In the past five decades, remarkable improvements have been made in surgical techniques, patient care, and treatment modalities5,6. However, the 5-year survival rate in surgically resected PDAC patients has barely risen to 17%. Nonetheless, this is still better than that in non-resected patients, which has remained almost unchanged (0.9%)4,7. Chemotherapy is the only other alternative PDAC treatment. Yet, this option is very limited as the great majority of PDAC patients exhibit strong resistance to chemotherapy medications such as Gemcitabine7,8. Other drugs, such as Erlotinib, are only available to a small group of PDAC patients with specific mutations, most of whom show Erlotinib resistance9. The adverse side effects associated with chemotherapy in most PDAC patients are yet another disadvantage of this treatment10. Recently, promising strategies have shown that immune checkpoint inhibitors (ICIs) and small molecule kinase inhibitors (SMKIs) can be effective in treating PDAC, but durable responses to these targeted therapies remain limited to a minority of patients11,12. Overall, the discovery of PDAC-specific early biomarkers can pave new avenues for early diagnosis and treatment.

PDAC develops from pancreatic intraepithelial neoplasms (PanIN) precursor lesions that result from non-invasive pancreatic duct epithelial proliferations13,14. While the formation of PanIN is initiated by oncogene mutations such as KRAS, additional genetic and epigenetic alterations are required for the progression to PDAC. It has been projected that the progression of PanIN through the different stages into invasive PDAC takes about 10 years13,15,16,17. This timeframe provides a great opportunity to benefit from early PDAC diagnosis. Therefore, extensive research has been carried out to establish tumor xenograft animal models and organoid cultures to study PDAC progression18,19,20,21. These models have been very useful for studying the invasive stages of PDAC, although not the transition from the early PanIN phases. It is, therefore, important to develop experimental models that can recapitulate the early progression of PanIN stages to enable the discovery of early detection biomarkers.

Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) using the four transcription factors OCT4, SOX2, KLF4, and c-MYC (OSKM) has illustrated the extent of cellular plasticity22. Cancer cell plasticity has been well-documented, and reprogramming human cancer cells into iPSCs has been successfully used to reset cells to their original cellular state, removing many of the epigenetic insults that have accumulated during cancer progression23,24,25,26,27,28,29. The possibility of using this reprogramming strategy to manipulate cancer cell identity has, therefore, presented great promise in treating cancer30,31. Indeed, we have previously shown that the differentiation of iPSCs derived from PDACs can recapitulate PDAC progression through the early PanIN stages32. By identifying genes and pathways specific to the early-to-intermediate stages of PDAC, candidate biomarkers were identified that can be clinically used for early PDAC diagnosis32,33. However, the biomarkers discovered using a single iPSC line showed limited coverage in the majority of PDAC patients32. The challenges of generating iPSC lines from other PDAC patients have halted the ability to discover more reliable biomarkers. This is due to many technical factors, including the heterogeneity of OSKM delivery, as only a small portion of human primary PDAC cells contained all four factors and responded successfully to reprogramming. Here, a detailed protocol is presented for reprogramming primary PDAC cells using a more efficient and consistent dual lentiviral delivery of OSKM.

Protocol

All experimental protocols were approved by the OHSU Institutional Review Board. All methods were carried out in accordance with relevant guidelines and regulations. All animal works for PDX tumors were performed with the OHSU Institutional Animal Use and Care Committee (IACUC) approval. This protocol was tested in Primary PDAC cells from patient-derived xenograft (PDX), BxPc3 cell line exhibiting epithelial morphology that was isolated from the pancreas tissue of a 61-year-old female patient with adenocarcinoma, the H6C…

Representative Results

Representative images displaying the morphology of iPSC colonies derived from PDAC, BXPc3, H6C7, and hFib cells are shown in Figure 1. PDAC-iPSC colonies started to form on Day 25 of reprogramming. Robust iPSC colonies with a more established ESC-like morphology were identified on Day 40 of reprogramming (Figure 1). Similarly, the formation of BxPc3-iPSCs began on Day 23 and became more established by Day 35. H6C7-iPSC formation …

Discussion

To facilitate the use of iPSC reprogramming for studying cancer progression, a robust protocol has been established for reprogramming pancreatic cancer cells. Reprogramming cancer cells into pluripotency has proven to be very challenging thus far, as only a few studies have successfully generated iPSCs from cancer cells32,36,37,38,39,<sup class="xre…

Divulgazioni

The authors have nothing to disclose.

Acknowledgements

A.S and J.K would like to thank Cancer Research UK and OHSU for funding (CRUK-OHSU Project Award C65925/A26986). A.S is supported by an MRC career development award (MR/N024028/1). A.A is funded by a Ph.D. scholarship (Scholarship ref. 1078107040) from King Abdulaziz City for Science and Technology. J.K is funded by MRF New Investigator Grant (GCNCR1042A) and Knight CEDAR grant (68182-933-000, 68182-939-000). We thank Prof Keisuke Kaji for kindly providing the reprogramming vector pSIN4-EF1a-O2S and pSIN4-CMV-K2M. For the purpose of open access, the author has applied a Creative Commons Attribution (CC BY) licence to any Author Accepted Manuscript version arising from this submission.

Materials

2-Mercaptoethanol (50 mM) Thermo Fisher 31350010
Alexa Fluor 488 anti- human TRA-1-60-R BioLegend 330613
Bovine Pituitary Extract (BPE) Thermo Fisher 13028014
BxPc3 ATCC CRL-1687
Cholera Toxin from Vibrio cholerae Merck  C8052-1MG
Collagen, Type I solution from rat tail Merck  C3867
Completed Defined K-SFM Thermo Fisher  10744-019
Corning Costar TC-Treated Multiple Well Plates Merck  CLS3516
Corning syringe filters Merck  CLS431231
Corning tissue-culture treated culture dishes Merck  CLS430599
Day Impex Virkon Disinfectant Virucidal Tablets Thermo Fisher 12328667
Dulbecco′s Phosphate Buffered Saline (PBS) Merck  D8537
Fetal Calf Serum (FCS)  Thermo Fisher 10270-106
Fugene HD Transfection Reagent  Promega   E2312
Gelatin solution, Type B, 2% in H2O Merck  G1393-100ML
Glasgow Minimum Essential Media (GMEM) Merck  G5154
Human EGF Recombinant Protein Thermo Fisher PHG0311
Human FGF-basic (FGF-2/bFGF) (154 aa) Recombinant Protein, PeproTech Thermo Fisher 100-18B
Human Pancreatic Duct Epithelial Cell Line (H6c7) Kerafast ECA001-FP
iMEF feeder cells  iXcells Biotechnologies 10MU-001-1V
Keratinocyte Serum Free Media (KSFM)  Thermo Fisher 17005-042
KnockOut DMEM  Thermo Fisher 10829018
KnockOut serum Replacement  Thermo Fisher 10828028
L-Glutamine (200 mM) Thermo Fisher 25030-024
MEM Non-Essential Amino Acids Solution (100x) Thermo Fisher 11140050
Millex-HP 0.45 μM syringe Filter Unit (Sterile) Merck  SLHP033RS
Opti-MEM Reduced Serum Medium  Thermo Fisher 31985062
pMDG  AddGene 187440
Polybrene (Hexadimethrine bromide)  Merck  H9268-5G
pSIN4-CMV-K2M  AddGene 21164
pSIN4-EF2-O2S  AddGene 21162
psPAX2 AddGene 12260
pWPT-GFP  AddGene 12255
RPMI 1640 Medium (ATCC modification) Thermo Fisher A1049101
Sodym Pyruvate Thermo Fisher 11360-039
Sterile Syringes for Single Use (60 mL)  Thermo Fisher 15899152
TrypLE Express Enzyme (1x), phenol red Thermo Fisher 12605036
UltraPure 0.5M EDTA, pH 8.0 Thermo Fisher 15575020
Y-27632 (Dihydrochloride) STEMCELL Technologies 72304

Riferimenti

  1. Rahib, L., et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Ricerca sul cancro. 74 (11), 2913-2921 (2014).
  2. Hu, J. X., et al. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World Journal of Gastroenterology. 27 (27), 4298 (2021).
  3. Howlader, N., et al. SEER cancer statistics review, 1975-2013. National Cancer Institute. 19, (2016).
  4. Bengtsson, A., Andersson, R., Ansari, D. The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data. Scientific Reports. 10 (1), 16425 (2020).
  5. He, J., et al. 2564 resected periampullary adenocarcinomas at a single institution: trends over three decades. HPB. 16 (1), 83-90 (2014).
  6. Dusch, N., et al. Factors predicting long-term survival following pancreatic resection for ductal adenocarcinoma of the pancreas: 40 years of experience. Journal of Gastrointestinal Surgery. 18 (4), 674-681 (2014).
  7. Principe, D. R., et al. The current treatment paradigm for pancreatic ductal adenocarcinoma and barriers to therapeutic efficacy. Frontiers in Oncology. 11, 688377 (2021).
  8. Papademetrio, D. L., et al. Interplay between autophagy and apoptosis in pancreatic tumors in response to gemcitabine. Targeted Oncology. 9 (2), 123-134 (2014).
  9. Ng, S. S., Tsao, M. S., Nicklee, T., Hedley, D. W. Effects of the epidermal growth factor receptor inhibitor OSI-774, Tarceva, on downstream signaling pathways and apoptosis in human pancreatic adenocarcinoma 1 supported by the National Cancer Institute of Canada and the Pat Myhal Fund for Pancreatic Cancer Research. Molecular Cancer Therapeutics. 1 (10), 777-783 (2002).
  10. Sultana, A., et al. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer. Journal of Clinical Oncology. 25 (18), 2607-2615 (2007).
  11. Sun, J., Russell, C. C., Scarlett, C. J., McCluskey, A. Small molecule inhibitors in pancreatic cancer. RSC Medicinal Chemistry. 11 (2), 164-183 (2020).
  12. Jiang, H., et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nature Medicine. 22 (8), 851-860 (2016).
  13. Hruban, R. H., et al. Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. The American Journal of Surgical Pathology. 25 (5), 579-586 (2001).
  14. Maitra, A., Hruban, R. H. Pancreatic cancer. Annual Review of Pathology: Mechanisms of Disease. 3, 157-188 (2008).
  15. Vincent, A., Herman, J., Schulick, R., Hruban, R. H., Goggins, M. Pancreatic cancer. The Lancet. 378 (9791), 607-620 (2011).
  16. Yachida, S., et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 467 (7319), 1114-1117 (2010).
  17. Lennon, A. M., et al. The early detection of pancreatic cancer: what will it take to diagnose and treat curable pancreatic neoplasia. Ricerca sul cancro. 74 (13), 3381-3389 (2014).
  18. Rubio-Viqueira, B., et al. An in vivo platform for translational drug development in pancreatic cancer. Clinical Cancer Research. 12 (15), 4652-4661 (2006).
  19. Li, C., et al. Identification of pancreatic cancer stem cells. Ricerca sul cancro. 67 (3), 1030-1037 (2007).
  20. Hermann, P. C., et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 1 (3), 313-323 (2007).
  21. Boj, S. F., et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 160 (1-2), 324-338 (2015).
  22. Takahashi, K., Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126 (4), 663-676 (2006).
  23. Suvà, M. L., et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell. 157, 580-594 (2014).
  24. Kotini, A. G., et al. Stage-specific human induced pluripotent stem cells map the progression of myeloid transformation to transplantable leukemia. Cell Stem Cell. 20 (3), 315-328 (2017).
  25. Stricker, S. H., et al. Widespread resetting of DNA methylation in glioblastoma-initiating cells suppresses malignant cellular behavior in a lineage-dependent manner. Genes Development. 27 (6), 654-669 (2013).
  26. Chao, M. P., et al. Human AML-iPSCs reacquire leukemic properties after differentiation and model clonal variation of disease. Cell Stem Cell. 20 (3), 329-344 (2017).
  27. Aparicio, L. A., et al. Clinical implications of epithelial cell plasticity in cancer progression. Cancer letters. 366 (1), 1-10 (2015).
  28. Grimont, A., Leach, S. D., Chandwani, R. Uncertain beginnings: acinar and ductal cell plasticity in the development of pancreatic cancer. Cellular and Molecular Gastroenterology and Hepatology. 13 (2), 369-382 (2022).
  29. Greenspan, L. J., Weinstein, B. M. To be or not to be: endothelial cell plasticity in development, repair, and disease. Angiogenesis. 24 (2), 251-269 (2021).
  30. Papapetrou, E. P. Patient-derived induced pluripotent stem cells in cancer research and precision oncology. Nature Medicine. 22 (12), 1392-1401 (2016).
  31. Zhang, X., Cruz, F. D., Terry, M., Remotti, F., Matushansky, I. Terminal differentiation and loss of tumorigenicity of human cancers via pluripotency-based reprogramming. Oncogene. 32 (18), 2249-2260 (2013).
  32. Kim, J., et al. An iPSC line from human pancreatic ductal adenocarcinoma undergoes early to invasive stages of pancreatic cancer progression. Cell Reports. 3 (6), 2088-2099 (2013).
  33. Kim, J., et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19-9 blood markers. Science Translational Medicine. 9 (398), (2017).
  34. Susac, L., et al. Structure of a fully assembled tumor-specific T cell receptor ligated by pMHC. Cell. 185 (17), 3201-3213 (2022).
  35. Yu, J., et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 324, 797-801 (2009).
  36. Carette, J. E., et al. Generation of iPSCs from cultured human malignant cells. Blood, The Journal of the American Society of Hematology. 115 (20), 4039-4042 (2010).
  37. Choi, S. M., et al. Reprogramming of EBV-immortalized B-lymphocyte cell lines into induced pluripotent stem cells. Blood, The Journal of the American Society of Hematology. 118 (7), 1801-1805 (2011).
  38. Hochedlinger, K., et al. Reprogramming of a melanoma genome by nuclear transplantation. Genes & Development. 18 (15), 1875-1885 (2004).
  39. Hu, K., et al. Efficient generation of transgene-free induced pluripotent stem cells from normal and neoplastic bone marrow and cord blood mononuclear cells. Blood, The Journal of the American Society of Hematology. 117 (14), e109-e119 (2011).
  40. Iskender, B., Izgi, K., Canatan, H. Reprogramming bladder cancer cells for studying cancer initiation and progression. Tumor Biology. 37, 13237-13245 (2016).
  41. Khoshchehreh, R., et al. Epigenetic reprogramming of primary pancreatic cancer cells counteracts their in vivo tumourigenicity. Oncogene. 38 (34), 6226-6239 (2019).
  42. Kim, H. J., et al. Establishment of hepatocellular cancer induced pluripotent stem cells using a reprogramming technique. Gut and Liver. 11 (2), 261 (2017).
  43. Lin, S. L., et al. Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. RNA. 14 (10), 2115-2124 (2008).
  44. Miyoshi, N., et al. Defined factors induce reprogramming of gastrointestinal cancer cells. Proceedings of the National Academy of Sciences. 107 (1), 40-45 (2010).
  45. Singovski, G., et al. In vivo epigenetic reprogramming of primary human colon cancer cells enhances metastases. Journal of Molecular Cell Biology. 8 (2), 157-173 (2016).
  46. Zhao, H., et al. A highly optimized protocol for reprogramming cancer cells to pluripotency using nonviral plasmid vectors. Cellular Reprogramming (Formerly" Cloning and Stem Cells&#34. 17 (1), 7-18 (2015).
  47. Lo, C. A., et al. Quantification of protein levels in single living cells). Cell Reports. 13 (11), 2634-2644 (2015).
  48. Yu, J., et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 324 (5928), 797-801 (2009).
  49. Prösch, S., et al. Inactivation of the very strong HCMV immediate early promoter by DNA CpG methylation in vitro. Biological Chemistry Hoppe-Seyler. 377 (3), 195-201 (1996).
  50. Mehta, A. K., Majumdar, S. S., Alam, P., Gulati, N., Brahmachari, V. Epigenetic regulation of cytomegalovirus major immediate-early promoter activity in transgenic mice. Gene. 428 (1-2), 20-24 (2009).
  51. Chen, X., et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell. 133 (6), 1106-1117 (2008).
  52. Kim, J., Chu, J., Shen, X., Wang, J., Orkin, S. H. An extended transcriptional network for pluripotency of embryonic stem cells. Cell. 132 (6), 1049-1061 (2008).
  53. Chronis, C., et al. Cooperative binding of transcription factors orchestrates reprogramming. Cell. 168 (3), 442-459 (2017).
  54. Li, D., et al. Chromatin accessibility dynamics during iPSC reprogramming. Cell Stem Cell. 21 (6), 819-833 (2017).
  55. Soufi, A., Donahue, G., Zaret, K. S. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell. 151 (5), 994-1004 (2012).
  56. Carey, B. W., et al. Reprogramming of murine and human somatic cells using a single polycistronic vector. Proceedings of the National Academy of Sciences of the United States of America. 106, 157-162 (2009).
check_url/it/65811?article_type=t

Play Video

Citazione di questo articolo
Alshaikh, A., Grygoryev, D., Keith, D., Sheppard, B., Sears, R. C., Kim, J., Soufi, A. Reprogramming Pancreatic Ductal Adenocarcinoma to Pluripotency. J. Vis. Exp. (204), e65811, doi:10.3791/65811 (2024).

View Video