CC BY 4.0 · TH Open 2019; 03(04): e340-e347
DOI: 10.1055/s-0039-1700885
Original Article
Georg Thieme Verlag KG Stuttgart · New York

Modelization of Blood-Borne Hypercoagulability in Myeloma: A Tissue-Factor-Bearing Microparticle-Driven Process

Loula Papageorgiou
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
2   Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
,
Kutaiba Alhaj Hussen
2   Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
3   INSERM U976, Université Paris-Diderot, École Pratique des Hautes Études/PSL Research University, Institut de recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
,
Sandrine Thouroude
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
,
Elisabeth Mbemba
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
,
Héléne Cost
4   Clinical Research, Diagnostica Stago, Gennevilliers, France
,
Laurent Garderet
5   Research Group “Proliferation and Differentiation of Stem Cells” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
,
Ismail Elalamy
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
2   Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
,
Annette Larsen
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
,
Patrick Van Dreden
4   Clinical Research, Diagnostica Stago, Gennevilliers, France
,
Meletios A. Dimopoulos
6   Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
,
Mohamad Mohty
7   Department of Hematology and Cell Therapy, Saint Antoine Hospital, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Sorbonne University, Paris, France
,
Grigoris T. Gerotziafas
1   Research Group “Cancer, Haemostasis and Angiogenesis,” INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Faculty of Medicine, Institut Universitaire de Cancérologie, Sorbonne Universities, Paris, France
2   Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
› Author Affiliations
Further Information

Publication History

16 April 2019

09 September 2019

Publication Date:
04 November 2019 (online)

Abstract

Introduction Hypercoagulability is a common blood alteration in newly diagnosed chemotherapy naïve patients with multiple myeloma. The identification of the procoagulant potential of cancer cells, which is principally related to tissue factor (TF) expression, attracts particular interest. The mechanisms by which myeloma plasma cells (MPCs) activate blood coagulation have been poorly investigated.

Aim To identify the principal actors related with MPCs that boost thrombin generation (TG).

Methods TF and annexin V expression by MPCs and MPC-derived microparticles (MPC-dMPs) was analyzed by flow cytometry. TF activity (TFa) and TF gene expression were also determined. TG in the presence of MPCs or MPC-dMPs was assessed with the calibrated automated thrombogram assay (CAT) in normal human PPP and in plasma depleted of factor VII or XII. TG was also assessed in plasma spiked with MPCs and MPC-dMPs.

Results MPC-dMPs expressed approximately twofold higher levels of TF as compared with MPCs. The TFa expressed by MPC-dMPs was significantly higher compared with that expressed by MPCs. MPCs and MPC-dMPs enhanced TG of human plasma. TG was significantly higher with MPC-dMPs compared with MPCs.

Conclusion MPCs indirectly induce blood-borne hypercoagulability through the release of MPC-dMPs rich in TF. Since MPCs, expressing low TFa, represent a weak procoagulant stimulus, the hypercoagulability at the microenvironment could be the resultant of MPC-dMPs rich in TF.

Authors' Contributions

L.P. did the experiments, has made substantial contributions to study design and organization, acquisition, analysis, and interpretation of data, and has been involved in drafting the manuscript. K.A.H. had substantial contribution in analysis and interpretation of the data from flow cytometry measurements. S.T. did the PCR experiments and interpreted the corresponding results. E.M. did the cultures of the cancer cells and prepared the samples with microparticles. H.C. was involved in the cultures of the cancer cells and thrombin generation experiments. L.G. contributed to the design of the study and the acquisition of the MPC cultures. I.E. was involved in the critical revision of the manuscript.


A.L. critically revised the manuscript. P.V.D. has made substantial contribution to the design of the study, the interpretation of the data, and the writing and editing of the manuscript. M.A.D. critically revised the manuscript. M.M. contributed to critical revision of the manuscript. G.T.G. as the principal investigator has made substantial contributions to conception and design of the study, analysis, and interpretation of data, was involved in drafting the manuscript, gave the final approval of the version to be published, and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.


 
  • References

  • 1 Fotiou D, Gerotziafas G, Kastritis E, Dimopoulos MA, Terpos E. A review of the venous thrombotic issues associated with multiple myeloma. Expert Rev Hematol 2016; 9 (07) 695-706
  • 2 Fotiou D, Sergentanis TN, Papageorgiou L. , et al. Longer procoagulant phospholipid-dependent clotting time, lower endogenous thrombin potential and higher tissue factor pathway inhibitor concentrations are associated with increased VTE occurrence in patients with newly diagnosed multiple myeloma: results of the prospective ROADMAP-MM-CAT study. Blood Cancer J 2018; 8 (11) 102
  • 3 Van Dreden P, Elalamy I, Gerotziafas GT. The role of tissue factor in cancer-related hypercoagulability, tumor growth, angiogenesis and metastasis and future therapeutic strategies. Crit Rev Oncog 2017; 22 (3–4): 219-248
  • 4 Undas A, Zubkiewicz-Usnarska L, Helbig G. , et al. Induction therapy alters plasma fibrin clot properties in multiple myeloma patients: association with thromboembolic complications. Blood Coagul Fibrinolysis 2015; 26 (06) 621-627
  • 5 Egyud LG, Lipinski B. Significance of fibrin formation and dissolution in the pathogenesis and treatment of cancer. Med Hypotheses 1991; 36 (04) 336-340
  • 6 Palumbo JS, Talmage KE, Massari JV. , et al. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005; 105 (01) 178-185
  • 7 Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer 2011; 11 (02) 123-134
  • 8 Egan K, Cooke N, Kenny D. Living in shear: platelets protect cancer cells from shear induced damage. Clin Exp Metastasis 2014; 31 (06) 697-704
  • 9 Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV. Thrombin-unique coagulation system protein with multifaceted impacts on cancer and metastasis. Cancer Metastasis Rev 2016; 35 (02) 213-233
  • 10 Falanga A, Schieppati F, Russo D. Cancer tissue procoagulant mechanisms and the hypercoagulable state of patients with cancer. Semin Thromb Hemost 2015; 41 (07) 756-764
  • 11 Gerotziafas GT, Galea V, Mbemba E. , et al. Tissue factor over-expression by human pancreatic cancer cells BXPC3 is related to higher prothrombotic potential as compared to breast cancer cells MCF7. Thromb Res 2012; 129 (06) 779-786
  • 12 Rousseau A, Larsen AK, Van Dreden P, Sabbah M, Elalamy I, Gerotziafas GT. Differential contribution of tissue factor and factor XII to thrombin generation triggered by breast and pancreatic cancer cells. Int J Oncol 2017; 51 (06) 1747-1756
  • 13 Marchetti M, Diani E, ten Cate H, Falanga A. Characterization of the thrombin generation potential of leukemic and solid tumor cells by calibrated automated thrombography. Haematologica 2012; 97 (08) 1173-1180
  • 14 Rousseau A, Van Dreden P, Khaterchi A, Larsen AK, Elalamy I, Gerotziafas GT. Procoagulant microparticles derived from cancer cells have determinant role in the hypercoagulable state associated with cancer. Int J Oncol 2017; 51 (06) 1793-1800
  • 15 Maacha S, Bhat AA, Jimenez L. , et al. Extracellular vesicles-mediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Mol Cancer 2019; 18 (01) 55
  • 16 Van Dreden P, Rousseau A, Savoure A, Lenormand B, Fontaine S, Vasse M. Plasma thrombomodulin activity, tissue factor activity and high levels of circulating procoagulant phospholipid as prognostic factors for acute myocardial infarction. Blood Coagul Fibrinolysis 2009; 20 (08) 635-641
  • 17 Butenas S, van't Veer C, Mann KG. “Normal” thrombin generation. Blood 1999; 94 (07) 2169-2178