Dendritic Cell-Based Immunotherapy in Advanced Sarcoma and Neuroblastoma Pediatric Patients: Anti-cancer Treatment Preceding Monocyte Harvest Impairs the Immunostimulatory and Antigen-Presenting Behavior of DCs and Manufacturing Process Outcome.
Journal: 2019/November - Frontiers in Oncology
ISSN: 2234-943X
Abstract:
Despite efforts to develop novel treatment strategies, refractory and relapsing sarcoma, and high-risk neuroblastoma continue to have poor prognoses and limited overall survival. Monocyte-derived dendritic cell (DC)-based anti-cancer immunotherapy represents a promising treatment modality in these neoplasias. A DC-based anti-cancer vaccine was evaluated for safety in an academic phase-I/II clinical trial for children, adolescents, and young adults with progressive, recurrent, or primarily metastatic high-risk tumors, mainly sarcomas and neuroblastomas. The DC vaccine was loaded with self-tumor antigens obtained from patient tumor tissue. DC vaccine quality was assessed in terms of DC yield, viability, immunophenotype, production of IL-12 and IL-10, and stimulation of allogenic donor T-cells and autologous T-cells in allo-MLR and auto-MLR, respectively. Here, we show that the outcome of the manufacture of DC-based vaccine is highly variable in terms of both DC yield and DC immunostimulatory properties. In 30% of cases, manufacturing resulted in a product that failed to meet medicinal product specifications and therefore was not released for administration to a patient. Focusing on the isolation of monocytes and the pharmacotherapy preceding monocyte harvest, we show that isolation of monocytes by elutriation is not superior to adherence on plastic in terms of DC yield, viability, or immunostimulatory capacity. Trial patients having undergone monocyte-interfering pharmacotherapy prior to monocyte harvest was associated with an impaired DC-based immunotherapy product outcome. Certain combinations of anti-cancer treatment resulted in a similar pattern of inadequate DC parameters, namely, a combination of temozolomide with irinotecan was associated with DCs showing poor maturation and decreased immunostimulatory features, and a combination of pazopanib, topotecan, and MTD-based cyclophosphamide was associated with poor monocyte differentiation and decreased DC immunostimulatory parameters. Searching for a surrogate marker predicting an adverse outcome of DC manufacture in the peripheral blood complete blood count prior to monocyte harvest, we observed an association between an increased number of immature granulocytes in peripheral blood and decreased potency of the DC-based product as quantified by allo-MLR. We conclude that the DC-manufacturing yield and the immunostimulatory quality of anti-cancer DC-based vaccines generated from the monocytes of patients were not influenced by the monocyte isolation modality but were detrimentally affected by the specific combination of anti-cancer agents used prior to monocyte harvest.
Relations:
Content
Citations
(3)
References
(39)
Diseases
(2)
Drugs
(7)
Chemicals
(2)
Anatomy
(4)
Affiliates
(3)
Similar articles
Articles by the same authors
Discussion board
Front Oncol 9: 1034

Dendritic Cell-Based Immunotherapy in Advanced Sarcoma and Neuroblastoma Pediatric Patients: Anti-cancer Treatment Preceding Monocyte Harvest Impairs the Immunostimulatory and Antigen-Presenting Behavior of DCs and Manufacturing Process Outcome

+8 authors

Supplementary Figure 1

Decision tree for DC-IMP manufacturing workflow including in-process and quality controls.

Click here for additional data file.(268K, pdf)

Supplementary Figure 2

Number of anti-cancer therapy lines preceding monocyte harvest were compared based on QC parameters: (A) DC yield, and post-thaw: (B) viability, (C) DC phenotype on day 0: CD14, CD197, CD80, CD86, CD83 and on day 2: MHC II, CD80, CD86, CD83 and immunostimulatory properties presented by (D) IL-12 production, IL-10 production and IL-12/IL-10 production ratio, (E) allo-MLR and auto-MLR.

Click here for additional data file.(842K, tif)

Supplementary Figure 3

Manufacturing subgroup from monocytes harvested after MTD-based therapy potentially interfering with monocyte biology and manufacturing subgroup from monocytes from untreated patients or after non-interfering treatment compared based on QC parameters: (A) DC yield, and post-thaw: (B) viability, (C) DC phenotype on day 0: CD14, CD197, CD80, CD86, CD83 and on day 2: MHC II, CD80, CD86, CD83 and immunostimulatory properties presented by (D) IL-12 production, IL-10 production and IL-12/IL-10 production ratio, (E) allo-MLR and auto-MLR.

Click here for additional data file.(857K, tif)

Supplementary Table 1

Monocyte biology-interfering medications.

Click here for additional data file.(65K, DOCX)

Supplementary Table 2

Study patient characteristics, disease course, and therapy.

Click here for additional data file.(85K, DOCX)

Supplementary Table 3

Source data: CBC parameters, manufacturing details, and QC parameters.

Click here for additional data file.(19K, XLSX)

Supplementary Material 1

html. Interactive—medications 60 days prior to monocyte harvest.

Click here for additional data file.(964K, ZIP)
Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
Department of Internal Medicine-Hematology and Oncology, University Hospital and Medical Faculty, Masaryk University, Brno, Czechia
Transfusion and Tissue Department, University Hospital Brno, Brno, Czechia
Edited by: Christina Annunziata, National Cancer Institute (NCI), United States
Reviewed by: Daniel Green, Kite Pharma, United States; Oladapo Yeku, Massachusetts General Hospital Cancer Center, United States
*Correspondence: Lenka Zdrazilova-Dubska zc.uom@aksbud
This article was submitted to Cancer Molecular Targets and Therapeutics, a section of the journal Frontiers in Oncology
†These authors have contributed equally to this work and are listed in alphabetical order
Edited by: Christina Annunziata, National Cancer Institute (NCI), United States
Reviewed by: Daniel Green, Kite Pharma, United States; Oladapo Yeku, Massachusetts General Hospital Cancer Center, United States
Received 2019 Jul 19; Accepted 2019 Sep 24.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

Abstract

Despite efforts to develop novel treatment strategies, refractory and relapsing sarcoma, and high-risk neuroblastoma continue to have poor prognoses and limited overall survival. Monocyte-derived dendritic cell (DC)-based anti-cancer immunotherapy represents a promising treatment modality in these neoplasias. A DC-based anti-cancer vaccine was evaluated for safety in an academic phase-I/II clinical trial for children, adolescents, and young adults with progressive, recurrent, or primarily metastatic high-risk tumors, mainly sarcomas and neuroblastomas. The DC vaccine was loaded with self-tumor antigens obtained from patient tumor tissue. DC vaccine quality was assessed in terms of DC yield, viability, immunophenotype, production of IL-12 and IL-10, and stimulation of allogenic donor T-cells and autologous T-cells in allo-MLR and auto-MLR, respectively. Here, we show that the outcome of the manufacture of DC-based vaccine is highly variable in terms of both DC yield and DC immunostimulatory properties. In 30% of cases, manufacturing resulted in a product that failed to meet medicinal product specifications and therefore was not released for administration to a patient. Focusing on the isolation of monocytes and the pharmacotherapy preceding monocyte harvest, we show that isolation of monocytes by elutriation is not superior to adherence on plastic in terms of DC yield, viability, or immunostimulatory capacity. Trial patients having undergone monocyte-interfering pharmacotherapy prior to monocyte harvest was associated with an impaired DC-based immunotherapy product outcome. Certain combinations of anti-cancer treatment resulted in a similar pattern of inadequate DC parameters, namely, a combination of temozolomide with irinotecan was associated with DCs showing poor maturation and decreased immunostimulatory features, and a combination of pazopanib, topotecan, and MTD-based cyclophosphamide was associated with poor monocyte differentiation and decreased DC immunostimulatory parameters. Searching for a surrogate marker predicting an adverse outcome of DC manufacture in the peripheral blood complete blood count prior to monocyte harvest, we observed an association between an increased number of immature granulocytes in peripheral blood and decreased potency of the DC-based product as quantified by allo-MLR. We conclude that the DC-manufacturing yield and the immunostimulatory quality of anti-cancer DC-based vaccines generated from the monocytes of patients were not influenced by the monocyte isolation modality but were detrimentally affected by the specific combination of anti-cancer agents used prior to monocyte harvest.

Keywords: dendritic cells, anti-cancer medications, sarcoma, neuroblastoma, cell-based medicinal products, investigator-initiated clinical trial, manufacturing outcome variability
Abstract

Footnotes

Funding. This work was supported by the Czech Ministry of Education, Youth and Sport via Large infrastructure CZECRIN (LM2015090) and RECAMO (LO1413), by the European Regional Development Fund—project CZECRIN_4PATIENTY (Reg. No. CZ.02.1.01/0.0/0.0/16_013/0001826), and by the Czech Ministry of Health via DRO 00209805.

Footnotes
Click here for additional data file.(268K, pdf)Click here for additional data file.(842K, tif)Click here for additional data file.(857K, tif)Click here for additional data file.(65K, DOCX)Click here for additional data file.(85K, DOCX)Click here for additional data file.(19K, XLSX)Click here for additional data file.(964K, ZIP)

References

  • 1. Oberlin O, Rey A, Lyden E, Bisogno G, Stevens MC, Meyer WH, et al. . Prognostic factors in metastatic rhabdomyosarcomas: results of a pooled analysis from United States and European cooperative groups. J Clin Oncol. (2008) 26:2384–9. 10.1200/JCO.2007.14.7207 ] [
  • 2. Pappo AS, Anderson JR, Crist WM, Wharam MD, Breitfeld PP, Hawkins D, et al. . Survival after relapse in children and adolescents with rhabdomyosarcoma: a report from the Intergroup Rhabdomyosarcoma Study Group. J Clin Oncol. (1999) 17:3487–93. 10.1200/JCO.1999.17.11.3487 [] [[PubMed]
  • 3. Sharp SE, Trout AT, Weiss BD, Gelfand MJ. MIBG in neuroblastoma diagnostic imaging and therapy. Radiographics. (2016) 36:258–78. 10.1148/rg.2016150099 [] [[PubMed]
  • 4. Zapletalova D, Andre N, Deak L, Kyr M, Bajciova V, Mudry P, et al. . Metronomic chemotherapy with the COMBAT regimen in advanced pediatric malignancies: a multicenter experience. Oncology. (2012) 82:249–60. 10.1159/000336483 [] [[PubMed]
  • 5. Berthold F, Homberg M, Proleskovskaya I, Mazanek P, Belogurova M, Ernst A, et al. . Metronomic therapy has low toxicity and is as effective as current standard treatment for recurrent high-risk neuroblastoma. Pediatr Hematol Oncol. (2017) 34:308–19. 10.1080/08880018.2017.1373314 [] [[PubMed]
  • 6. Krishnadas DK, Shusterman S, Bai F, Diller L, Sullivan JE, Cheerva AC, et al. . A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma. Cancer Immunol Immunother. (2015) 64:1251–60. 10.1007/s00262-015-1731-3 [] [[PubMed]
  • 7. Merchant MS, Bernstein D, Amoako M, Baird K, Fleisher TA, Morre M, et al. . Adjuvant immunotherapy to improve outcome in high-risk pediatric sarcomas. Clin Cancer Res. (2016) 22:3182–91. 10.1158/1078-0432.CCR-15-2550 [] [[PubMed]
  • 8. Klement GL, Arkun K, Valik D, Roffidal T, Hashemi A, Klement C, et al. . Future paradigms for precision oncology. Oncotarget. (2016) 7:46813–31. 10.18632/oncotarget.9488 ] [
  • 9. Rossi M, Young JW. Human dendritic cells: potent antigen-presenting cells at the crossroads of innate and adaptive immunity. J Immunol. (2005) 175:1373–81. 10.4049/jimmunol.175.3.1373 [] [[PubMed]
  • 10. Hoffman R, Benz EJ, Jr, Shattil S, Furie B, Cohen H. Hematology: Basic Principles and Practice. Churchill Livingstone; (2005). [PubMed]
  • 11. Jakubzick CV, Randolph GJ, Henson PM. Monocyte differentiation and antigen-presenting functions. Nat Rev Immunol. (2017) 17:349–62. 10.1038/nri.2017.28 [] [[PubMed]
  • 12. Boyette LB, Macedo C, Hadi K, Elinoff BD, Walters JT, Ramaswami B, et al. . Phenotype, function, and differentiation potential of human monocyte subsets. PLoS ONE. (2017) 12:e0176460. 10.1371/journal.pone.0176460 ] [
  • 13. Patel AA, Zhang Y, Fullerton JN, Boelen L, Rongvaux A, Maini AA, et al. . The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med. (2017) 214:1913–23. 10.1084/jem.20170355 ] [
  • 14. Sampath P, Moideen K, Ranganathan UD, Bethunaickan R. Monocyte subsets: phenotypes and function in tuberculosis infection. Front Immunol. (2018) 9:1726. 10.3389/fimmu.2018.01726 ] [
  • 15. Zhou JX, Feng LJ, Zhang X. Risk of severe hematologic toxicities in cancer patients treated with PARP inhibitors: a meta-analysis of randomized controlled trials. Drug Des Devel Ther. (2017) 11:3009–17. 10.2147/DDDT.S147726 ] [
  • 16. Rusten LS, Lyman SD, Veiby OP, Jacobsen SE. The FLT3 ligand is a direct and potent stimulator of the growth of primitive and committed human CD34+ bone marrow progenitor cells in vitro. Blood. (1996) 87:1317–25. [[PubMed]
  • 17. Kao J, Timmins J, Ozao-Choy J, Packer S. Effects of combined sunitinib and extracranial stereotactic radiotherapy on bone marrow hematopoiesis. Oncol Lett. (2016) 12:2139–44. 10.3892/ol.2016.4851 ] [
  • 18. Briegert M, Kaina B. Human monocytes, but not dendritic cells derived from them, are defective in base excision repair and hypersensitive to methylating agents. Cancer Res. (2007) 67:26–31. 10.1158/0008-5472.CAN-06-3712 [] [[PubMed]
  • 19. Bauer M, Goldstein M, Heylmann D, Kaina B. Human monocytes undergo excessive apoptosis following temozolomide activating the ATM/ATR pathway while dendritic cells and macrophages are resistant. PLoS ONE. (2012) 7:e39956. 10.1371/journal.pone.0039956 ] [
  • 20. Dijkgraaf EM, Heusinkveld M, Tummers B, Vogelpoel LT, Goedemans R, Jha V, et al. . Chemotherapy alters monocyte differentiation to favor generation of cancer-supporting M2 macrophages in the tumor microenvironment. Cancer Res. (2013) 73:2480–92. 10.1158/0008-5472.CAN-12-3542 [] [[PubMed]
  • 21. Hu J, Kinn J, Zirakzadeh AA, Sherif A, Norstedt G, Wikstrom AC, et al. . The effects of chemotherapeutic drugs on human monocyte-derived dendritic cell differentiation and antigen presentation. Clin Exp Immunol. (2013) 172:490–9. 10.1111/cei.12060 ] [
  • 22. R Core Team . R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna: (2019). [PubMed]
  • 23. Ge C, Li R, Song H, Geng T, Yang J, Tan Q, et al. . Phase I clinical trial of a novel autologous modified-DC vaccine in patients with resected NSCLC. BMC Cancer. (2017) 17:884. 10.1186/s12885-017-3859-3 ] [
  • 24. Rodriguez J, Castanon E, Perez-Gracia JL, Rodriguez I, Viudez A, Alfaro C, et al. . A randomized phase II clinical trial of dendritic cell vaccination following complete resection of colon cancer liver metastasis. J Immunother Cancer. (2018) 6:96. 10.1186/s40425-018-0405-z ] [
  • 25. Repnik U, Knezevic M, Jeras M. Simple and cost-effective isolation of monocytes from buffy coats. J Immunol Methods. (2003) 278:283–92. 10.1016/S0022-1759(03)00231-X [] [[PubMed]
  • 26. Dohnal AM, Graffi S, Witt V, Eichstill C, Wagner D, Ul-Haq S, et al. . Comparative evaluation of techniques for the manufacturing of dendritic cell-based cancer vaccines. J Cell Mol Med. (2009) 13:125–35. 10.1111/j.1582-4934.2008.00304.x ] [
  • 27. Zdrazilova Dubska L, Fedorova L, Pilatova K, Mudry P, Hlavackova E, Matoulkova E, et al TKI pazopanib impaires immunostimulatory properties of monocytes: implication for monocyte-derived DC-based anti-cancer vaccine preparation. Ann Oncol. (2016) 27:18P 10.1093/annonc/mdw525.18 [[PubMed][Google Scholar]
  • 28. Trojandt S, Knies D, Pektor S, Ritz S, Mailander V, Grabbe S, et al. . The chemotherapeutic agent topotecan differentially modulates the phenotype and function of dendritic cells. Cancer Immunol Immunother. (2013) 62:1315–26. 10.1007/s00262-013-1431-9 [] [[PubMed]
  • 29. Cai W, Maldonado NV, Cui W, Harutyunyan N, Ji L, Sposto R, et al. . Activity of irinotecan and temozolomide in the presence of O6-methylguanine-DNA methyltransferase inhibition in neuroblastoma pre-clinical models. Br J Cancer. (2010) 103:1369–79. 10.1038/sj.bjc.6605927 ] [
  • 30. Seitz M, Zwicker M, Loetscher P. Effects of methotrexate on differentiation of monocytes and production of cytokine inhibitors by monocytes. Arthritis Rheum. (1998) 41:2032–8. 10.1002/1529-0131(199811)41:11<2032::AID-ART19>3.0.CO;2-J [] [[PubMed]
  • 31. Cutolo M, Sulli A, Craviotto C, Felli L, Pizzorni C, Seriolo B, et al. . Antiproliferative-antiinflammatory effects of methotrexate and sex hormones on cultured differentiating myeloid monocytic cells (THP-1). Ann N Y Acad Sci. (2002) 966:232–7. 10.1111/j.1749-6632.2002.tb04220.x [] [[PubMed]
  • 32. Moller B, Kukoc-Zivojnov N, Okamgba S, Kessler U, Puccetti E, Ottmann OG, et al. . Folinic acid antagonizes methotrexate-induced differentiation of monocyte progenitors. Rheumatol Int. (2002) 22:60–7. 10.1007/s00296-002-0188-9 [] [[PubMed]
  • 33. Perpetuo IP, Caetano-Lopes J, Rodrigues AM, Campanilho-Marques R, Ponte C, Canhao H, et al. . Methotrexate and low-dose prednisolone downregulate osteoclast function by decreasing receptor activator of nuclear factor-kappabeta expression in monocytes from patients with early rheumatoid arthritis. RMD Open. (2017) 3:e000365. 10.1136/rmdopen-2016-000365 ] [
  • 34. Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol. (2014) 14:302–14. 10.1038/nri3660 [] [[PubMed]
  • 35. Pilatova K, Bencsikova B, Demlova R, Valik D, Zdrazilova-Dubska L. Myeloid-derived suppressor cells (MDSCs) in patients withsolid tumors: considerations for granulocyte colony-stimulating factor treatment. Cancer Immunol Immunother. (2018) 67:1919–29. 10.1007/s00262-018-2166-4 [] [[PubMed]
  • 36. Hartung T, Docke WD, Gantner F, Krieger G, Sauer A, Stevens P, et al. . Effect of granulocyte colony-stimulating factor treatment on ex vivo blood cytokine response in human volunteers. Blood. (1995) 85:2482–9. [[PubMed]
  • 37. Mielcarek M, Graf L, Johnson G, Torok-Storb B. Production of interleukin-10 by granulocyte colony-stimulating factor-mobilized blood products: a mechanism for monocyte-mediated suppression of T-cell proliferation. Blood. (1998) 92:215–22. [[PubMed]
  • 38. Boneberg EM, Hareng L, Gantner F, Wendel A, Hartung T. Human monocytes express functional receptors for granulocyte colony-stimulating factor that mediate suppression of monokines and interferon-gamma. Blood. (2000) 95:270–6. [[PubMed]
  • 39. Saito M, Kiyokawa N, Taguchi T, Suzuki K, Sekino T, Mimori K, et al. . Granulocyte colony-stimulating factor directly affects human monocytes and modulates cytokine secretion. Exp Hematol. (2002) 30:1115–23. 10.1016/S0301-472X(02)00889-5 [] [[PubMed]
  • 40. Hori S, Heike Y, Takei M, Maruyama M, Inoue Y, Lee JJ, et al. . Freeze-thawing procedures have no influence on the phenotypic and functional development of dendritic cells generated from peripheral blood CD14+ monocytes. J Immunother. (2004) 27:27–35. 10.1097/00002371-200401000-00003 [] [[PubMed]
  • 41. Ghanekar SA, Bhatia S, Ruitenberg JJ, Dela Rosa C, Disis ML, Maino VC, et al. . Phenotype and in vitro function of mature MDDC generated from cryopreserved PBMC of cancer patients are equivalent to those from healthy donors. J Immune Based Ther Vaccines. (2007) 5:7. 10.1186/1476-8518-5-7 ] [
  • 42. Silveira GF, Wowk PF, Machado AM, Duarte Dos Santos CN, Bordignon J. Immature dendritic cells generated from cryopreserved human monocytes show impaired ability to respond to LPS and to induce allogeneic lymphocyte proliferation. PLoS ONE. (2013) 8:e71291. 10.1371/journal.pone.0071291 ] [
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.