REFERENCES

1. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014;74:2913-21.

2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7-33.

3. Al-Share B, Hammad N, Diab M. Pancreatic adenocarcinoma: molecular drivers and the role of targeted therapy. Cancer Metastasis Rev 2021;40:355-71.

4. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817-25.

5. Singh RR, O’Reilly EM. New treatment strategies for metastatic pancreatic ductal adenocarcinoma. Drugs 2020;80:647-69.

6. Väyrynen SA, Zhang J, Yuan C, et al. Composition, spatial characteristics, and prognostic significance of myeloid cell infiltration in pancreatic cancer. Clin Cancer Res 2021;27:1069-81.

7. Park W, Chawla A, O’Reilly EM. Pancreatic cancer: a review. JAMA 2021;326:851-62.

8. Vaish U, Jain T, Are AC, Dudeja V. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma: an update on heterogeneity and therapeutic targeting. Int J Mol Sci 2021;22:13408.

9. Gopurappilly R, Bhat V, Bhonde R. Pancreatic tissue resident mesenchymal stromal cell (MSC)-like cells as a source of in vitro islet neogenesis. J Cell Biochem 2013;114:2240-7.

10. Quante M, Tu SP, Tomita H, et al. Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell 2011;19:257-72.

11. Mathew E, Brannon AL, Del Vecchio A, et al. Mesenchymal stem cells promote pancreatic tumor growth by inducing alternative polarization of macrophages. Neoplasia 2016;18:142-51.

12. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res 2007;67:10123-8.

13. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest 2002;110:341-50.

14. Özdemir BC, Pentcheva-Hoang T, Carstens JL, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014;25:719-34.

15. Chen Y, Kim J, Yang S, et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021;39:548-565.e6.

16. Bhattacharjee S, Hamberger F, Ravichandra A, et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J Clin Invest 2021;131:146987.

17. Lee JJ, Perera RM, Wang H, et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci USA 2014;111:E3091-100.

18. Walter K, Omura N, Hong SM, et al. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin Cancer Res 2010;16:1781-9.

19. Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol 2015;33:4284-92.

20. Elyada E, Bolisetty M, Laise P, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov 2019;9:1102-23.

21. Baron M, Veres A, Wolock SL, et al. A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure. Cell Syst 2016;3:346-360.e4.

22. Bernard V, Semaan A, Huang J, et al. Single-cell transcriptomics of pancreatic cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. Clin Cancer Res 2019;25:2194-205.

23. Dominguez CX, Müller S, Keerthivasan S, et al. Single-cell RNA sequencing reveals stromal evolution into LRRC15+ myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov 2020;10:232-53.

24. Hosein AN, Huang H, Wang Z, et al. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight 2019;5:129212.

25. Öhlund D, Handly-Santana A, Biffi G, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med 2017;214:579-96.

26. Biffi G, Oni TE, Spielman B, et al. IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov 2019;9:282-301.

27. Van Cutsem E, Tempero MA, Sigal D, et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J Clin Oncol 2020;38:3185-94.

28. Ramanathan RK, McDonough SL, Philip PA, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol 2019;37:1062-9.

29. Kemp SB, Pasca di Magliano M, Crawford HC. Myeloid cell mediated immune suppression in pancreatic cancer. Cell Mol Gastroenterol Hepatol 2021;12:1531-42.

30. Sharma V, Aggarwal A, Jacob J, Sahni D. Myeloid-derived suppressor cells: bridging the gap between inflammation and pancreatic adenocarcinoma. Scand J Immunol 2021;93:e13021.

31. Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 2016;7:12150.

32. Vanhaver C, van der Bruggen P, Bruger AM. MDSC in mice and men: mechanisms of immunosuppression in cancer. J Clin Med 2021;10:2872.

33. Trovato R, Fiore A, Sartori S, et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J Immunother Cancer 2019;7:255.

34. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest 2015;125:3365-76.

35. Huang B, Pan PY, Li Q, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res 2006;66:1123-31.

36. Zhang Y, Velez-Delgado A, Mathew E, et al. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017;66:124-36.

37. Chao T, Furth EE, Vonderheide RH. CXCR2-dependent accumulation of tumor-associated neutrophils regulates T-cell immunity in pancreatic ductal adenocarcinoma. Cancer Immunol Res 2016;4:968-82.

38. Bayne LJ, Beatty GL, Jhala N, et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012;21:822-35.

39. Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012;21:836-47.

40. Stromnes IM, Brockenbrough JS, Izeradjene K, et al. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut 2014;63:1769-81.

41. Steele CW, Karim SA, Leach JDG, et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell 2016;29:832-45.

42. Yang S, Liu Q, Liao Q. Tumor-associated macrophages in pancreatic ductal adenocarcinoma: origin, polarization, function, and reprogramming. Front Cell Dev Biol 2020;8:607209.

43. Sanford DE, Belt BA, Panni RZ, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 2013;19:3404-15.

44. Zhu Y, Herndon JM, Sojka DK, et al. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity 2017;47:323-338.e6.

45. Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res 2007;67:9518-27.

46. Ziske C, Etzrodt PE, Eliu AS, et al. Increase of in vivo antitumoral activity by CD40L (CD154) gene transfer into pancreatic tumor cell-dendritic cell hybrids. Pancreas 2009;38:758-65.

47. Liu Q, Li Y, Niu Z, et al. Atorvastatin (Lipitor) attenuates the effects of aspirin on pancreatic cancerogenesis and the chemotherapeutic efficacy of gemcitabine on pancreatic cancer by promoting M2 polarized tumor associated macrophages. J Exp Clin Cancer Res 2016;35:33.

48. Poh AR, Ernst M. Tumor-associated macrophages in pancreatic ductal adenocarcinoma: therapeutic opportunities and clinical challenges. Cancers 2021;13:2860.

49. Candido JB, Morton JP, Bailey P, et al. CSF1R+ macrophages sustain pancreatic tumor growth through T Cell suppression and maintenance of key gene programs that define the squamous subtype. Cell Rep 2018;23:1448-60.

50. Zhu Y, Knolhoff BL, Meyer MA, et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res 2014;74:5057-69.

51. Wang-gillam A, O’reilly EM, Bendell JC, et al. A randomized phase II study of cabiralizumab (cabira) + nivolumab (nivo) ± chemotherapy (chemo) in advanced pancreatic ductal adenocarcinoma (PDAC). JCO 2019;37:TPS465.

52. Nywening TM, Wang-gillam A, Sanford DE, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol 2016;17:651-62.

53. Noel M, O’Reilly EM, Wolpin BM, et al. Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs 2020;38:800-11.

54. Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011;331:1612-6.

55. Beatty GL, Winograd R, Evans RA, et al. Exclusion of T cells from pancreatic carcinomas in mice is regulated by Ly6C(low) F4/80(+) extratumoral macrophages. Gastroenterology 2015;149:201-10.

56. Winograd R, Byrne KT, Evans RA, et al. Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol Res 2015;3:399-411.

57. O’hara MH, O’reilly EM, Varadhachary G, et al. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: an open-label, multicentre, phase 1b study. Lancet Oncol 2021;22:118-31.

58. Lee JW, Stone ML, Porrett PM, et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 2019;567:249-52.

59. Nielsen SR, Quaranta V, Linford A, et al. Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nat Cell Biol 2016;18:549-60.

60. Lohneis P, Sinn M, Bischoff S, et al. Cytotoxic tumour-infiltrating T lymphocytes influence outcome in resected pancreatic ductal adenocarcinoma. Eur J Cancer 2017;83:290-301.

61. Steele NG, Carpenter ES, Kemp SB, et al. Multimodal mapping of the tumor and peripheral blood immune landscape in human pancreatic cancer. Nat Cancer 2020;1:1097-112.

62. Topcu KSB, Korucu EN, Menevse E, Kocak N, Duran T. Investigation of the effects of the toll-like receptor 4 pathway on immune checkpoint vista in pancreatic cancer. Invest New Drugs 2022;40:519-28.

63. Hou Z, Pan Y, Fei Q, et al. Prognostic significance and therapeutic potential of the immune checkpoint VISTA in pancreatic cancer. J Cancer Res Clin Oncol 2021;147:517-31.

64. Frey N, Tortola L, Egli D, et al. Loss of Rnf31 and Vps4b sensitizes pancreatic cancer to T cell-mediated killing. Nat Commun 2022;13:1804.

65. Tian C, Huang Y, Clauser KR, et al. Suppression of pancreatic ductal adenocarcinoma growth and metastasis by fibrillar collagens produced selectively by tumor cells. Nat Commun 2021;12:2328.

66. Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer 2020;20:174-86.

67. Akce M, Shaib WL, Diab M, et al. Phase Ib/II trial of siltuximab and spartalizumab in patients in metastatic pancreatic cancer. JCO 2022;40:TPS626-TPS626.

68. Bockorny B, Semenisty V, Macarulla T, et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med 2020;26:878-85.

69. Seo YD, Jiang X, Sullivan KM, et al. Mobilization of CD8+ T cells via CXCR4 blockade facilitates PD-1 checkpoint therapy in human pancreatic cancer. Clin Cancer Res 2019;25:3934-45.

70. Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA 2013;110:20212-7.

71. Goulart MR, Stasinos K, Fincham REA, Delvecchio FR, Kocher HM. T cells in pancreatic cancer stroma. World J Gastroenterol 2021;27:7956-68.

72. Diana A, Wang LM, D’Costa Z, et al. Prognostic value, localization and correlation of PD-1/PD-L1, CD8 and FOXP3 with the desmoplastic stroma in pancreatic ductal adenocarcinoma. Oncotarget 2016;7:40992-1004.

73. Tang Y, Xu X, Guo S, et al. An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma. PLoS One 2014;9:e91551.

74. Zhang Y, Lazarus J, Steele NG, et al. Regulatory T-cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis. Cancer Discov 2020;10:422-39.

75. McAllister F, Bailey JM, Alsina J, et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 2014;25:621-37.

76. Zhang Y, Zoltan M, Riquelme E, et al. Immune cell production of interleukin 17 Induces stem cell features of pancreatic intraepithelial neoplasia cells. Gastroenterology 2018;155:210-223.e3.

77. Barilla RM, Diskin B, Caso RC, et al. Specialized dendritic cells induce tumor-promoting IL-10+IL-17+ FoxP3neg regulatory CD4+ T cells in pancreatic carcinoma. Nat Commun 2019;10:1424.

78. Pylayeva-Gupta Y, Das S, Handler JS, et al. IL35-producing B cells promote the development of pancreatic neoplasia. Cancer Discov 2016;6:247-55.

79. Roghanian A, Fraser C, Kleyman M, Chen J. B Cells promote pancreatic tumorigenesis. Cancer Discov 2016;6:230-2.

80. Castino GF, Cortese N, Capretti G, et al. Spatial distribution of B cells predicts prognosis in human pancreatic adenocarcinoma. Oncoimmunology 2016;5:e1085147.

81. Minici C, Rigamonti E, Lanzillotta M, et al. B lymphocytes contribute to stromal reaction in pancreatic ductal adenocarcinoma. Oncoimmunology 2020;9:1794359.

82. Tong DN, Guan J, Sun JH, et al. Characterization of B cell-mediated PD-1/PD-L1 interaction in pancreatic cancer patients. Clin Exp Pharmacol Physiol 2020;47:1342-9.

83. Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, Fallon PG. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun 2015;6:5997.

84. Gunderson AJ, Kaneda MM, Tsujikawa T, et al. Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov 2016;6:270-85.

85. Tempero M, Oh DY, Tabernero J, et al. Ibrutinib in combination with nab-paclitaxel and gemcitabine for first-line treatment of patients with metastatic pancreatic adenocarcinoma: phase III RESOLVE study. Ann Oncol 2021;32:600-8.

86. Philip PA. Targeting B cells in pancreatic adenocarcinoma: does RESOLVE resolve the question? Ann Oncol 2021;32:582-3.

87. Byrne KT, Vonderheide RH. CD40 stimulation obviates innate sensors and drives T cell immunity in cancer. Cell Rep 2016;15:2719-32.

88. Ma HS, Poudel B, Torres ER, et al. A CD40 agonist and PD-1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T-cell-mediated anticancer activity. Cancer Immunol Res 2019;7:428-42.

89. Byrne KT, Betts CB, Mick R, et al. Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer. Clin Cancer Res 2021;27:4574-86.

90. Yang J. Dendritic cells in pancreatic cancer immunotherapy: vaccines and combination immunotherapies. Pathol Res Pract 2019;215:152691.

91. Yang J, Shangguan J, Eresen A, Li Y, Wang J, Zhang Z. Dendritic cells in pancreatic cancer immunotherapy: vaccines and combination immunotherapies. Pathol Res Pract 2019;215:152691.

92. Hirooka S. The role of circulating dendritic cells in patients with unresectable pancreatic cancer. Anticancer Res 2011;31:3827-34.

93. Yamamoto T, Yanagimoto H, Satoi S, et al. Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. J Surg Res 2012;173:299-308.

94. Fukunaga A, Miyamoto M, Cho Y, et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 2004;28:e26-31.

95. Dallal RM, Christakos P, Lee K, Egawa S, Son YI, Lotze MT. Paucity of dendritic cells in pancreatic cancer. Surgery 2002;131:135-8.

96. Baylor college of medicine, N. Th-1 Dendritic cell immunotherapy plus standard chemotherapy for pancreatic adenocarcinoma (DECIST). Available from: https://clinicaltrials.gov/ct2/show/NCT04157127 [Last accessed on 27 September 2022].

97. Katsuda M, Miyazawa M, Ojima T, et al. A double-blind randomized comparative clinical trial to evaluate the safety and efficacy of dendritic cell vaccine loaded with WT1 peptides (TLP0-001) in combination with S-1 in patients with advanced pancreatic cancer refractory to standard chemotherapy. Trials 2019;20:242.

98. Nagai K, Adachi T, Harada H, Eguchi S, Sugiyama H, Miyazaki Y. Dendritic cell-based immunotherapy pulsed with wilms tumor 1 peptide and mucin 1 as an adjuvant therapy for pancreatic ductal adenocarcinoma after curative resection: a phase I/IIa clinical trial. Anticancer Res 2020;40:5765-76.

99. Hoshikawa M, Aoki T, Matsushita H, et al. NK cell and IFN signatures are positive prognostic biomarkers for resectable pancreatic cancer. Biochem Biophys Res Commun 2018;495:2058-65.

100. Lim SA, Kim J, Jeon S, et al. Defective localization with impaired tumor cytotoxicity contributes to the immune escape of NK cells in pancreatic cancer patients. Front Immunol 2019;10:496.

101. Dickson I. Pancreatic cancer: γδ T cells support PDAC. Nat Rev Gastroenterol Hepatol 2016;13:560.

102. Seifert AM, List J, Heiduk M, et al. Gamma-delta T cells stimulate IL-6 production by pancreatic stellate cells in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2020;146:3233-40.

103. Chiossone L, Dumas PY, Vienne M, Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol 2018;18:671-88.

104. Moral JA, Leung J, Rojas LA, et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 2020;579:130-5.

Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/