Next Article in Journal
Rheumatoid Synovial Fluid and Acidic Extracellular pH Modulate the Immunomodulatory Activity of Urine-Derived Stem Cells
Next Article in Special Issue
Editorial: Multiple Myeloma: Molecular Mechanism and Targeted Therapy
Previous Article in Journal
A Comparative Study between Onion Peel Extracts, Free and Complexed with β-Cyclodextrin, as a Natural UV Filter to Cosmetic Formulations
Previous Article in Special Issue
Genetic Abnormalities in Extramedullary Multiple Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems

by
Anushka Kulkarni
1,
Despina Bazou
2 and
Maria José Santos-Martinez
1,3,4,*
1
The School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, The University of Dublin, D02 PN40 Dublin, Ireland
2
School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
3
School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
4
Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(21), 15855; https://doi.org/10.3390/ijms242115855
Submission received: 27 September 2023 / Revised: 25 October 2023 / Accepted: 29 October 2023 / Published: 1 November 2023
(This article belongs to the Special Issue Multiple Myeloma: Molecular Mechanism and Targeted Therapy)

Abstract

:
Multiple myeloma (MM) is a hematological malignancy originated in the bone marrow and characterized by unhindered plasma cell proliferation that results in several clinical manifestations. Although the main role of blood platelets lies in hemostasis and thrombosis, platelets also play a pivotal role in a number of other pathological conditions. Platelets are the less-explored components from the tumor microenvironment in MM. Although some studies have recently revealed that MM cells have the ability to activate platelets even in the premalignant stage, this phenomenon has not been widely investigated in MM. Moreover, thrombocytopenia, along with bleeding, is commonly observed in those patients. In this review, we discuss the hemostatic disturbances observed in MM patients and the dynamic interaction between platelets and myeloma cells, along with present and future potential avenues for the use of platelets for diagnostic and therapeutic purposes.

1. Multiple Myeloma: Clinical Manifestations and Treatment

Multiple myeloma (MM) is a bone marrow malignancy that accounts for 1% of all cancer types. An average of 80 thousand people are diagnosed with MM in Europe and the United States every year [1,2,3] and it is more common in people above 60 years of age, particularly in men [4].
The disease is characterized by the unhindered proliferation of B cells in the bone marrow. It can affect several parts of the body including, but not limited to, the spine, ribs, kidney, blood, and brain, leading to several clinical manifestations including hypercalcemia, renal failure, anemia, and bone pain due to the presence of lytic lesions (CRAB). The diagnostic criteria for active MM include the presence of more than 60% of clonal bone marrow plasma cells, a serum free light chain ratio (FLC) ≥ 100, and the presence of at least one focal lesion of 5 mm or greater identified by magnetic resonance imaging (MRI) [5].
MM usually begins as an asymptomatic premalignant condition known as monoclonal gammopathy of unknown significance (MGUS) and smoldering multiple myeloma (SMM). Patients with MGUS and SMM usually have excessive circulating quantities of monoclonal antibodies, and elevated serum and urine microglobulin levels without the presence of clinical symptoms [6,7]. SMM is associated with a higher level of infiltrating plasma cells (from 10% to less than 60%) and higher microglobulin levels in serum and urine. Progression of MGUS (less than 10% of infiltrating plasma cells) to symptomatic MM is low, 1% per year, while the percentage of patients that progress to MM among SMM patients is ten times higher [8]. The progression toward MM is associated with the presence of various molecular markers [9]. The International Staging System for MM takes into consideration factors such as the gradual acquisition of cytogenetic abnormalities (translocations, insertion/deletion of chromosomes), the number of plasma cells in the bone marrow (BMPC), serum and urine M- protein levels, serum FLC ratio, and β2 microglobulin and albumin levels as criteria for establishing the patient stage (Table 1).
Various molecular markers have been shown to confer poor prognosis. These include the deletion of chromosome 13q, 17p [11,12], and the presence of increased levels of serum beta macroglobulin and LDH [13].
The International Myeloma Foundation and European Hematology Association (EHA) and the European Society for Medical Oncology (ESMO) have laid down guidelines for the management of MM patients, based on published data from clinical trials [14,15]. For newly diagnosed patients, triplet therapy, including an immunomodulatory drug (IMID), e.g., lenalidomide, a proteasome inhibitor (PI), e.g., bortezomib, and the corticosteroid dexamethasone, is preferred, followed by autologous stem cell transplantation. For second-line treatment, a combination of different IMIDs and PIs is implemented followed by PI-based regimens [16]. Immunotherapies with monoclonal antibodies like daratumumab, elotuzumab, and isatuximab are also included in the treatment plan. Recently, bispecifics have shown great responses in relapsed/refractory MM patients with one to three prior therapies. The US-FDA and European Commission conditionally approved Talquetamab, marketed by Janssen under the name Talvey®. Talquetamab is a bispecific antibody, the first in its class to target the novel receptor GPRC5D (NCT04634552) [17]. Similarly, both Janssen’s Teclistamab and Pfizer’s Elrexfio (elranatamab-bcmm) bispecific antibodies target the B-cell maturation antigen (BCMA)-cluster of differentiation three (CD3) (NCT04557098, NCT04649359) in relapsed/refractory MM patients [18,19].

2. Hemostatic Complications Associated with Multiple Myeloma

Cancer-associated thrombosis (CAT) is frequently observed in cancer patients. In fact, venous thromboembolism (VTE) is commonly associated with disease progression and increased mortality. CAT has a complex underlying mechanism that has been associated with the secretion of tissue factor (TF), increased platelet activation, and endothelial cell dysfunction, among others [20]. VTE is commonly presented in patients with MM, even at the initial stages of MGUS [21]. VTE risk is especially higher within the first year of MGUS and active disease diagnosis. However, VTE is not an individual risk factor for the progression of MGUS to active MM [22]. Risk-predicting models (IMPEDE [23], SAVED [24], and PRISM score [25]) can be used for predicting the risk of VTE development in MM patients. However, extensive validation of these models is required before clinical use. Kapur et al. [26] showed that none of the models accurately predicted the VTE risk of MM patients and many patients in the intermediate and low-risk groups still developed VTE.
While the mechanisms underlying thrombosis have been extensively explored in solid tumors, these are still under-studied in MM. Cancer cells in solid tumors contribute to thrombosis by secreting TF and cancer procoagulant (CP). TF is constitutively expressed in different cancers such as leukemia, breast, pancreatic, brain, prostate, and ovarian cancer, apart from being secreted by monocytes and endothelial cells [27,28,29,30,31]. TF has been associated with the development of VTE in ovarian cancer patients [30]; however, no such correlation was found in patients with brain tumors [31]. TF can also bind to and activate factor VII, thus activating the coagulation cascade [32]. CP is a serine protease found in some cancers such as acute myeloid leukemia and Lewis lung carcinoma, and can directly activate factor X, thus contributing to the hypercoagulable state of the disease [20,33,34]. In MM, increased levels of thrombin generation, vWF (which is also elevated in solid tumors), and fibrin polymerization have been employed as potential indicators of thrombosis. Van Marion et al. argued that these alone do not contribute to the development of VTE, rather, patient- and treatment-specific factors should be taken into account [35,36]. In addition, Ghansah et al. suggested that resistance to activated protein C (APC) can lead to a hypercoagulable MM state as APC did not attenuate thrombin generation in MM patients [37]. More recently, Nielson et al. argued that EVs from MM patients exert procoagulant activity by inducing thrombin generation and carrying TF and procoagulant phospholipids. The researchers also found that this activity diminished during the course of treatment with induction therapy consisting of bortezomib, cyclophosphamide, and dexamethasone [38].
On the other hand, bleeding is also occasionally seen in MM patients. When compared to individuals with solid tumor malignancies, patients with hematological cancers are more likely to experience thrombocytopenia. Patients with MM have been shown to have the highest rates among those with hematological malignancies [39,40]. In addition to thrombocytopenia, dysfibrinogenemia brought on by the interaction of numerous paraproteins with the proteins in the coagulation cascade can also contribute to bleeding in MM patients [41,42,43,44].
Interestingly, there is a recent study by Li et al. [45] that compared the thrombin generation profile in whole blood and poor platelet plasma of 21 MM patients with healthy controls to understand the paradoxical existence of both bleeding and thrombosis in MM. They used a new method to measure thrombin generation in whole blood that takes into account the potential involvement of platelets and red and white blood cells during this process. The authors concluded in their work that “patients balance on a thin line between pro and anti-coagulant phenotype” and hypothesize that both platelets (thrombocytopenia and platelet dysfunction) and red blood cells (that promote thrombin generation in vivo) play a crucial role in this balance.

3. Hemostatic Complications Associated with Multiple Myeloma Treatment

While the current therapeutic agents used for treating MM have consistently demonstrated a tolerable safety profile with significant and clinically relevant benefits, thrombosis and bleeding in MM patients can also be attributed to chemotherapy.
Treatment with immunomodulatory agents like lenalidomide and dexamethasone has increased the prevalence of VTE in MM. A two-to-four-fold increased platelet activity in lenalidomide-treated MM patients has been observed, as assessed by an increased aggregation response and P-selectin upregulation. A significant increase in coagulation activator markers when compared to controls (patients at diagnosis) was also reported [46,47,48].
Bortezomib has been shown to induce reversible thrombocytopenia, probably due to the inhibition of platelet formation from megakaryocytes and an increase in the expression of GTPase Rho. [49] Although some patients on bortezomib may require platelet transfusion [50], concomitant therapy with high doses of dexamethasone has been reported to reduce the need for transfusions [51]. An in vitro study carried out on platelet-rich plasma from healthy human volunteers has also shown that bortezomib can inhibit ADP-induced platelet aggregation [52]. In three extremely rare cases, bortezomib has also been associated with severe diffuse alveolar hemorrhage in MM patients [53].
Daratumumab, an antibody that targets CD38 and induces direct antimyeloma activity, has been associated with a higher incidence of thrombocytopenia [54].
Guidelines recommend using aspirin or low-molecular-weight heparins (LMWHs) in MM patients [55]. Bleeding can also be associated with anticoagulant therapy. However, a study carried out on 1605 patients on warfarin (45.7%), LMWHs (29%), and direct orally acting anticoagulants (25.3%) demonstrated that only a small percentage (3.9%) of patients suffered from bleeding during a follow-up period of 1.6 years [56].

4. Platelet–Myeloma Cell Interactions

Platelets play a dynamic role in the bone marrow microenvironment by interacting with other cells locally. Their interaction with hematopoietic stem cells can occur through the release of platelet-derived growth factor (PDGF), transforming growth factor β-1 (TGF-β1), and vascular endothelial growth factor (VEGF), all of them involved in the cell growth and survival of hematopoietic cells [57]. Platelets and leukocytes also interact with each other by physical contact and via the release of chemical mediators [58,59]. For instance, the binding of platelets to monocytes via P-selectin (CD62P) leads to the release of the platelet-activating factor and TF, resulting in the activation of more platelets [60].
There are limited studies investigating the interaction between platelets and myeloma cells. In 2018, Takagi et al. showed that several MM cell lines (MM.1S, KMS-11, U266, OPM-2, and H929) were able to induce platelet aggregation [61]. In addition, when these cell lines were co-cultured with platelets or platelet releasate from healthy donors, cell proliferation was significantly enhanced, except for MM.1S. The authors then injected OPM-2 cells pre-exposed to platelets into mice and observed a high rate of tumor cell engraftment that resulted in decreased mice survival. They attributed this effect to IL-1β upregulation as the IL-1β OPM-2 knockout cells abrogated tumor engraftment [61].
MM cells release TNF-α [62] which is a potent platelet activator [63]. There are, however, contradictory results concerning platelet activation in the premalignant MGUS state and active MM disease. In the premalignant MGUS stage, the upregulation of P-selectin on the platelet surface and the abundance of plasma-soluble P-selectin levels have been reported [48,64,65]. On the other hand, platelets from patients with active MM have shown impaired platelet activation as evaluated by P-selectin expression in response to several agonists including collagen, ADP, epinephrine, and protease-activated receptor-1 (PAR-1)-activating peptide [66]. In another study, there was no significant difference in P-selectin expression between healthy donors and MM patients at diagnosis [46]. Myeloma cells can also produce IL-6 [67]. Apart from inhibiting apoptosis in MM cells [68] and increasing RANKL [69], IL-6 can increase the platelet count by increasing thrombopoietin production [70]. It is therefore imperative that further studies are required to elucidate the role of platelets in the different MM disease stages.
Given the limited number of studies exploring the molecular interactions between MM cells and platelets, we propose that this interaction could take place through several mechanisms, as depicted in Figure 1. MM cells express platelet-derived growth factor receptor (PDGFR) α and β which could bind PDGF released by activated platelets [71]. The upregulation of this receptor is also strongly correlated to angiogenesis and microvessel density (MVD) [72]. P-selectin glycoprotein ligand-1 (PSGL-1) expressed on MM cells could bind P-selectin on activated platelets. Leukocytes and myeloid cells also express PSGL-1 that could bind to activated platelets [73]. It has been reported that MM patients with no PSGL-1 expression had a lower survival; however, a potential mechanism behind this was not reported [74]. Syndecan-1 (CD138) is also expressed on the surface of MM cells and when shed, it promotes tumor growth and metastasis. Syndecan-1 can also bind to the angiogenic factors, VEGF, and fibroblast growth factor-2 (FGF-2), promoting angiogenesis and negatively affecting patient prognosis [75,76].
Platelet factor-4 (PF4) is stored in the platelet alpha granules and released upon activation. In MM patient serum samples, a downregulation of PF4 has been reported [77]. However, human recombinant PF4 has shown proapoptotic activity by inhibiting MM cell proliferation and angiogenesis, through the inhibition of the STAT3 and IL6-STAT3 pathways, both in vitro and in vivo [78]. Platelets also release sphingosine-1 phosphate (S1P) that acts as a tumorigenic and angiogenic growth factor. In myeloma, S1P binds to SIP1, SIP2, and SIP3 receptors on myeloma plasma cells, preventing dexamethasone-induced apoptosis [79,80]

5. Platelets as Diagnostic and Therapeutic Tools in Cancer

Cells have been engineered to be used as drug delivery vehicles due to their extended stability, minimal immunogenicity, and selective targeting [81]. Utilizing platelets as drug delivery vehicles appears to be a promising alternative since they have been implicated in the proliferation and spread of tumor cells.
Platelet microvesicles (PMVs) are released by activated platelets. Microvesicles are extracellular vesicles (EVs) that range from 100 to 1000 nm and are released by the outward blebbing of the cell membrane. It is intriguing that despite the platelets’ absence of a nucleus, PMVs may transport genetic material like RNA, mRNA, and microRNA. PMVs can interact with tumor cells as well as circulating immune cells via receptors such as CXCR4, which aids in the growth and spread of tumors [82,83], and GPIIb/IIIa, which enhances tumor survival and migration [84]. PMVs also contain lipid mediators, such as eicosanoids, which may be utilized as biomarkers [85]. Furthermore, they are nearly 100 times more procoagulant than platelets due to their higher prothrombotic potential [86]. Due to their small size, they can enter tissues more readily. Recently, due to their excellent stability and low immunogenicity, platelet EVs (PEVs) have demonstrated the potential to be employed as drug carriers. Both hydrophilic and hydrophobic drugs have been successfully loaded into PEVs, mainly through two methods: (a) platelet incubation with drugs and (b) drug loading directly into EVs through sonication, freeze–thaw cycles, dialysis, and electroporation. [86,87].
In addition to PMVs [88], platelets themselves can also be employed as drug carriers because they interact with tumor cells via tumor cell-induced platelet aggregation (TCIPA), promoting the passive delivery of the drug to the tumor site [89]. This approach has been successful in preliminary studies where platelets have been loaded with doxorubicin and subsequently delivered to lymphoma cells in vitro [90]. In this study, doxorubicin was loaded through the open canalicular system with a high loading and encapsulation efficiency. In vivo studies using athymic BALB/c-nude mice have demonstrated the ability of the doxorubicin-loaded platelets to accumulate at the tumor site. In addition, a reduced accumulation of doxorubicin-loaded platelets in the heart tissue was also observed, highlighting that via this approach, cardiotoxic side effects associated with doxorubicin can be minimized [90]. Furthermore, platelets’ surface can be functionalized with various moieties that could actively target tumor cell receptors. An antibody against programmed death ligand-1 (PD-L1) is gaining a lot of attention to selectively capture residual circulatory tumor cells (CTCs) [91]. Platelets coated with anti-PD-L1 were engineered by Gu and his colleagues to reduce, after resection, tumor recurrence in triple-negative breast carcinoma and melanoma in a mouse model and demonstrated that they successfully inhibited tumor recurrence when compared to controls [92]. In another study by Hu et al. [93], platelets bound to anti-PD-L1 and conjugated to hematopoietic stem cells were intravenously administered in a C1498 leukemia mouse model to target tumor cells. The conjugated system migrated to the bone marrow and released anti-PD-L1, thus increasing T-cell migration and cytokine production, resulting in increased survival. This is extremely interesting as platelets could be further employed to bispecifically target the aggregation process along with the inclusion of a chemotherapeutic agent. Coating nanoparticles with platelet membranes is also an attractive option as these nanoparticles can actively target the site of platelet action [94]. According to in vivo experiments performed on mice, platelet membrane-coated polymeric nanoparticles loaded with the anti-MM agent bortezomib and tissue plasminogen activator were able to selectively target MM in the bone marrow [95]. The design of ”human nanoplatelets” with a tunable surface and adequate loading capacity by Dai et al. represents a novel approach for the selective targeting and imaging of early-stage tumors, as they have shown in RPMI8226-derived MM xenotransplants in NOD/SCID mice models [96].
More recently, platelets have been shown to take up bioactive factors secreted by tumor cells, including mRNA from CTCs. The genetic makeup of platelets can potentially be altered by CTCs. Such platelets are called tumor-educated platelets (TEPs) [97]. TEPs could therefore be exploited as a cutting-edge biomarker for early disease detection and intervention. While TEPs have been employed as biomarkers for the early diagnosis of primary thyroid cancer, glioblastoma, ovarian cancer, and renal cell carcinoma [98,99,100,101], their role in hematological malignancies and MM specifically has not been explored.

6. Conclusions

The role of platelets in MM has not been extensively investigated. Several multiple myeloma cell lines, including MM.1S, U266, KMS-11, OPM-2, and H929, have been shown to induce platelet aggregation. Additionally, platelets and platelet releasate have been shown to stimulate tumor cell proliferation in vitro, increase tumor engraftment, and decrease mice survival. Regarding platelet activation, although additional research is required, studies in patients have demonstrated increased platelet activation in the premalignant MGUS stage and a lowered activation in the active disease stage. Hemostatic disturbances are frequently observed in newly diagnosed MM patients, and they are also associated with disease treatment. Given the role that platelets play in tumor progression and metastasis, the use of platelets as vehicles has gained popularity in recent years. ”human nanoplatelets” and ”platelet membrane coated nanoparticles” with a tunable surface area are explored for diagnosis and to target tumor cells. Such approaches would be of great potential for the early detection of MM, including the premalignant stages, as well as in the disease management.

Author Contributions

A.K. conceived and wrote the manuscript. M.J.S.-M. and D.B. conceived, reviewed, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

AK is a holder of a Postgraduate Ussher Fellowship from Trinity College Dublin, The University of Dublin, Ireland. DB is funded by grant HRCI-HRB-2020-022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Multiple Myeloma Source: Globocan 2020. Available online: https://gco.iarc.fr/today/data/factsheets/cancers/35-Multiple-myeloma-fact-sheet.pdf (accessed on 5 November 2022).
  2. Multiple myeloma|Irish Cancer Society. Available online: https://www.cancer.ie/cancer-information-and-support/cancer-types/multiple-myeloma (accessed on 5 November 2022).
  3. Key Statistics for Multiple Myeloma. Available online: https://www.cancer.org/cancer/multiple-myeloma/about/key-statistics.html (accessed on 2 April 2023).
  4. Incidence of Myeloma—Myeloma Patients Europe. Available online: https://www.mpeurope.org/about-myeloma/incidence-of-myeloma/ (accessed on 5 November 2022).
  5. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group Updated Criteria for the Diagnosis of Multiple Myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef] [PubMed]
  6. Waldenstrom, J. Studies on Conditions Associated with Disturbed Gamma Globulin Formation (Gammopathies). Harvey Lect. 1960, 56, 211–231. [Google Scholar] [PubMed]
  7. Moser-Katz, T.; Joseph, N.S.; Dhodapkar, M.V.; Lee, K.P.; Boise, L.H. Game of Bones: How Myeloma Manipulates Its Microenvironment. Front. Oncol. 2020, 10, 625199. [Google Scholar] [CrossRef] [PubMed]
  8. Rajkumar, S.V. MGUS and Smoldering Multiple Myeloma: Update on Pathogenesis, Natural History, and Management. Hematology 2005, 2005, 340–345. [Google Scholar] [CrossRef] [PubMed]
  9. Dima, D.; Jiang, D.; Singh, D.J.; Hasipek, M.; Shah, H.S.; Ullah, F.; Khouri, J.; Maciejewski, J.P.; Jha, B.K. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers 2022, 14, 4082. [Google Scholar] [CrossRef] [PubMed]
  10. International Staging System for Multiple Myeloma | The IMF. Available online: https://www.myeloma.org/international-staging-system-iss-reivised-iss-r-iss (accessed on 14 May 2023).
  11. Avet-Loiseau, H.; Attal, M.; Moreau, P.; Charbonnel, C.; Garban, F.; Hulin, C.; Leyvraz, S.; Michallet, M.; Yakoub-Agha, I.; Garderet, L.; et al. Genetic Abnormalities and Survival in Multiple Myeloma: The Experience of the Intergroupe Francophone Du Myélome. Blood 2007, 109, 3489–3495. [Google Scholar] [CrossRef]
  12. Flynt, E.; Bisht, K.; Sridharan, V.; Ortiz, M.; Towfic, F.; Thakurta, A. Prognosis, Biology, and Targeting of TP53 Dysregulation in Multiple Myeloma. Cells 2020, 9, 287. [Google Scholar] [CrossRef]
  13. Wallington-Beddoe, C.T.; Mynott, R.L. Prognostic and Predictive Biomarker Developments in Multiple Myeloma. J. Hematol. Oncol. 2021, 14, 151. [Google Scholar] [CrossRef]
  14. Multiple Myeloma Treatment Overview | Int’l Myeloma Foundation. Available online: https://www.myeloma.org/multiple-myeloma-treatment (accessed on 1 September 2023).
  15. Dimopoulos, M.A.; Moreau, P.; Terpos, E.; Mateos, M.V.; Zweegman, S.; Cook, G.; Delforge, M.; Hájek, R.; Schjesvold, F.; Cavo, M.; et al. Multiple Myeloma: EHA-ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2021, 32, 309–322. [Google Scholar] [CrossRef]
  16. Goldman-Mazur, S.; Visram, A.; Rajkumar, S.V.; Kapoor, P.; Dispenzieri, A.; Lacy, M.Q.; Gertz, M.A.; Buadi, F.K.; Hayman, S.R.; Dingli, D.; et al. Second Line Treatment Strategies in Multiple Myeloma: A Referral-Center Experience. Blood 2021, 138 (Suppl. S1), 819. [Google Scholar] [CrossRef]
  17. A Study of Talquetamab in Participants With Relapsed or Refractory Multiple Myeloma—Full Text View—ClinicalTrials.gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04634552 (accessed on 28 August 2023).
  18. MagnetisMM-3: Study Of Elranatamab (PF-06863135) Monotherapy in Participants With Multiple Myeloma Who Are Refractory to at Least One PI, One IMiD and One Anti-CD38 mAb—Full Text View—ClinicalTrials.gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04649359 (accessed on 30 August 2023).
  19. Moreau, P.; Garfall, A.L.; van de Donk, N.W.C.J.; Nahi, H.; San-Miguel, J.F.; Oriol, A.; Nooka, A.K.; Martin, T.; Rosinol, L.; Chari, A.; et al. Teclistamab in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2022, 387, 495–505. [Google Scholar] [CrossRef] [PubMed]
  20. Mukai, M.; Oka, T. Mechanism and Management of Cancer-Associated Thrombosis. J. Cardiol. 2018, 72, 89–93. [Google Scholar] [CrossRef] [PubMed]
  21. Eby, C. Pathogenesis and Management of Bleeding and Thrombosis in Plasma Cell Dyscrasias. Br. J. Haematol. 2009, 145, 151–163. [Google Scholar] [CrossRef] [PubMed]
  22. Kristinsson, S.Y.; Fears, T.R.; Gridley, G.; Turesson, I.; Mellqvist, U.H.; Björkholm, M.; Landgren, O. Deep Vein Thrombosis after Monoclonal Gammopathy of Undetermined Significance and Multiple Myeloma. Blood 2008, 112, 3582–3586. [Google Scholar] [CrossRef] [PubMed]
  23. Sanfilippo, K.M.; Luo, S.; Wang, T.-F.; Wildes, T.; Mikhael, J.; Keller, J.W.; Thomas, T.S.; Carson, K.R.; Gage, B.F. Predicting Risk of Venous Thromboembolism in Multiple Myeloma: The Impede VTE Score. Blood 2018, 132 (Suppl. S1), 141. [Google Scholar] [CrossRef]
  24. Li, A.; Wu, Q.; Luo, S.; Warnick, G.S.; Zakai, N.A.; Libby, E.N.; Gage, B.F.; Garcia, D.A.; Lyman, G.H.; Sanfilippo, K.M. Derivation and Validation of a Risk Assessment Model for Immunomodulatory Drug–Associated Thrombosis Among Patients With Multiple Myeloma. J. Natl. Compr. Cancer Netw. 2019, 17, 840–847. [Google Scholar] [CrossRef] [PubMed]
  25. Chakraborty, R.; Rybicki, L.; Wei, W.; Valent, J.; Faiman, B.M.; Samaras, C.J.; Anwer, F.; Khorana, A.A. Abnormal Metaphase Cytogenetics Predicts Venous Thromboembolism in Myeloma: Derivation and Validation of the PRISM Score. Blood 2022, 140, 2443–2450. [Google Scholar] [CrossRef]
  26. Kapur, S.; Feehan, K.; Mosiman, S.; Frankki, S.; Rosenstein, L.J. Real World Validation of VTE Risk Models in Newly Diagnosed Multiple Myeloma in a Community Setting. Blood 2021, 138 (Suppl. S1), 2971. [Google Scholar] [CrossRef]
  27. Thaler, J.; Ay, C.; Mackman, N.; Metz-Schimmerl, S.; Stift, J.; Kaider, A.; Müllauer, L.; Gnant, M.; Scheithauer, W.; Pabinger, I. Microparticle-Associated Tissue Factor Activity in Patients with Pancreatic Cancer: Correlation with Clinicopathological Features. Eur. J. Clin. Invest. 2013, 43, 277–285. [Google Scholar] [CrossRef]
  28. Lwaleed, B.A.; Lam, L.; Lasebai, M.; Cooper, A.J. Expression of Tissue Factor and Tissue Factor Pathway Inhibitor in Microparticles and Subcellular Fractions of Normal and Malignant Prostate Cell Lines. Blood Coagul. Fibrinolysis 2013, 24, 339–343. [Google Scholar] [CrossRef]
  29. Ueno, T.; Toi, M.; Koike, M.; Nakamura, S.; Tominaga, T. Tissue Factor Expression in Breast Cancer Tissues: Its Correlation with Prognosis and Plasma Concentration. Br. J. Cancer 2000, 83, 164–170. [Google Scholar] [CrossRef] [PubMed]
  30. Uno, K.; Homma, S.; Satoh, T.; Nakanishi, K.; Abe, D.; Matsumoto, K.; Oki, A.; Tsunoda, H.; Yamaguchi, I.; Nagasawa, T.; et al. Tissue Factor Expression as a Possible Determinant of Thromboembolism in Ovarian Cancer. Br. J. Cancer 2007, 96, 290. [Google Scholar] [CrossRef] [PubMed]
  31. Thaler, J.; Preusser, M.; Ay, C.; Kaider, A.; Marosi, C.; Zielinski, C.; Pabinger, I.; Hainfellner, J.A. Intratumoral Tissue Factor Expression and Risk of Venous Thromboembolism in Brain Tumor Patients. Thromb. Res. 2013, 131, 162–165. [Google Scholar] [CrossRef] [PubMed]
  32. Kasthuri, R.S.; Taubman, M.B.; Mackman, N. Role of Tissue Factor in Cancer. J. Clin. Oncol. 2009, 27, 4834. [Google Scholar] [CrossRef] [PubMed]
  33. Hilgard, P.; Whur, P. Factor X-Activating Activity from Lewis Lung Carcinoma. Br. J. Cancer 1980, 41, 642–643. [Google Scholar] [CrossRef] [PubMed]
  34. Falanga, A.; Alessio, M.; Donati, M.; Barbui, T. A New Procoagulant in Acute Leukemia. Blood 1988, 71, 870–875. [Google Scholar] [CrossRef]
  35. van Marion, A.M.W.; Auwerda, J.J.A.; Lisman, T.; Sonneveld, P.; de Maat, M.P.M.; Lokhorst, H.M.; Leebeek, F.W.G. Prospective Evaluation of Coagulopathy in Multiple Myeloma Patients before, during and after Various Chemotherapeutic Regimens. Leuk. Res. 2008, 32, 1078–1084. [Google Scholar] [CrossRef]
  36. Fotiou, D.; Gavriatopoulou, M.; Terpos, E. Multiple Myeloma and Thrombosis: Prophylaxis and Risk Prediction Tools. Cancers 2020, 12, 191. [Google Scholar] [CrossRef]
  37. Ghansah, H.; Debreceni, I.B.; Váróczy, L.; Rejtő, L.; Lóczi, L.; Bagoly, Z.; Kappelmayer, J. Patients with Multiple Myeloma and Monoclonal Gammopathy of Undetermined Significance Have Variably Increased Thrombin Generation and Different Sensitivity to the Anticoagulant Effect of Activated Protein C. Thromb. Res. 2023, 223, 44–52. [Google Scholar] [CrossRef]
  38. Nielsen, T.; Kristensen, S.R.; Gregersen, H.; Teodorescu, E.M.; Christiansen, G.; Pedersen, S. Extracellular Vesicle-Associated Procoagulant Phospholipid and Tissue Factor Activity in Multiple Myeloma. PLoS ONE 2019, 14, e0210835. [Google Scholar] [CrossRef]
  39. Shaw, J.L.; Nielson, C.M.; Park, J.K.; Marongiu, A.; Soff, G.A. The Incidence of Thrombocytopenia in Adult Patients Receiving Chemotherapy for Solid Tumors or Hematologic Malignancies. Eur. J. Haematol. 2021, 106, 662–672. [Google Scholar] [CrossRef] [PubMed]
  40. Bennett, D.; Suppapanya, N.; Grotzinger, K. Thrombocytopenia in Hematologic Malignancy and Solid Tumors in the United States. J. Clin. Oncol. 2012, 30 (Suppl. S15), e12001. [Google Scholar] [CrossRef]
  41. Rahman, S.; Veeraballi, S.; Chan, K.H.; Shaaban, H.S. Bleeding Diathesis in Multiple Myeloma: A Rare Presentation of a Dreadful Emergency with Management Nightmare. Cureus 2021, 13, e13990. [Google Scholar] [CrossRef] [PubMed]
  42. Siddiq, N.; Bergstrom, C.; Anderson, L.D.; Nagalla, S. Bleeding Due to Acquired Dysfibrinogenemia as the Initial Presentation of Multiple Myeloma. BMJ Case Rep. CP 2019, 12, e229312. [Google Scholar] [CrossRef] [PubMed]
  43. Shinagawa, A.; Kojima, H.; Berndt, M.C.; Kaneko, S.; Suzukawa, K.; Hasegawa, Y.; Shigeta, O.; Nagasawa, T. Characterization of a Myeloma Patient with a Life-Threatening Hemorrhagic Diathesis: Presence of a Lambda Dimer Protein Inhibiting Shear-Induced Platelet Aggregation by Binding to the A1 Domain of von Willebrand Factor. Thromb. Haemost. 2005, 93, 889–896. [Google Scholar] [CrossRef]
  44. Saif, M.W.; Allegra, C.J.; Greenberg, B. Bleeding Diathesis in Multiple Myeloma. J. Hematother. Stem. Cell Res. 2001, 10, 657–660. [Google Scholar] [CrossRef]
  45. Li, L.; Roest, M.; Sang, Y.; Remijn, J.A.; Fijnheer, R.; Smit, K.; Huskens, D.; Wan, J.; de Laat, B.; Konings, J. Patients with Multiple Myeloma Have a Disbalanced Whole Blood Thrombin Generation Profile. Front. Cardiovasc. Med. 2022, 9, 919495. [Google Scholar] [CrossRef]
  46. Gibbins, J.; Rana, R.; Khan, D.; Shapiro, S.; Grech, H.; Ramasamy, K. Multiple Myeloma Treatment Is Associated with Enhanced Platelet Reactivity. Blood 2018, 132 (Suppl. S1), 3300. [Google Scholar] [CrossRef]
  47. Knight, R.; DeLap, R.J.; Zeldis, J.B. Lenalidomide and Venous Thrombosis in Multiple Myeloma. N. Engl. J. Med. 2006, 354, 2079–2080. [Google Scholar] [CrossRef]
  48. Robak, M.; Treliński, J.; Chojnowski, K. Hemostatic Changes after 1 Month of Thalidomide and Dexamethasone Therapy in Patients with Multiple Myeloma. Med. Oncol. 2012, 29, 3574–3580. [Google Scholar] [CrossRef]
  49. Murai, K.; Kowata, S.; Shimoyama, T.; Yashima-Abo, A.; Fujishima, Y.; Ito, S.; Ishida, Y. Bortezomib Induces Thrombocytopenia by the Inhibition of Proplatelet Formation of Megakaryocytes. Eur. J. Haematol. 2014, 93, 290–296. [Google Scholar] [CrossRef] [PubMed]
  50. Lonial, S.; Waller, E.K.; Richardson, P.G.; Jagannath, S.; Orlowski, R.Z.; Giver, C.R.; Jaye, D.L.; Francis, D.; Giusti, S.; Torre, C.; et al. Risk Factors and Kinetics of Thrombocytopenia Associated with Bortezomib for Relapsed, Refractory Multiple Myeloma. Blood 2005, 106, 3777. [Google Scholar] [CrossRef] [PubMed]
  51. Quach, H.; Prince, M.H.; Honemann, D.; Westerman, D.; Milner, A.D.; Barron, A.; Harrison, S. High-Dose Dexamethasone Reduces Bortezomib-Induced Thrombocytopenia. Blood 2007, 110, 4820. [Google Scholar] [CrossRef]
  52. Avcu, F.; Ural, A.U.; Cetin, T.; Nevruz, O. Effects of Bortezomib on Platelet Aggregation and ATP Release in Human Platelets, In Vitro. Thromb. Res. 2008, 121, 567–571. [Google Scholar] [CrossRef]
  53. Ayed, A.O.; Moreb, J.S.; Hsu, J.W.; Hiemenz, J.W.; Wingard, J.R.; Norkin, M. Severe Diffuse Alveolar Hemorrhage Associated with Bortezomib Administration in Patients with Multiple Myeloma. Blood 2014, 124, 5761. [Google Scholar] [CrossRef]
  54. Palumbo, A.; Chanan-Khan, A.; Weisel, K.; Nooka, A.K.; Masszi, T.; Beksac, M.; Spicka, I.; Hungria, V.; Munder, M.; Mateos, M.V.; et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2016, 375, 754–766. [Google Scholar] [CrossRef] [PubMed]
  55. Palumbo, A.; Rajkumar, S.V.; Dimopoulos, M.A.; Richardson, P.G.; San Miguel, J.; Barlogie, B.; Harousseau, J.; Zonder, J.A.; Cavo, M.; Zangari, M.; et al. Prevention of Thalidomide- and Lenalidomide-Associated Thrombosis in Myeloma. Leukemia 2008, 22, 414–423. [Google Scholar] [CrossRef]
  56. Adrianzen Herrera, D.; Lutsey, P.L.; Giorgio, K.; Zakai, N.A. Bleeding Risk in Patients with Multiple Myeloma Treated for Venous Thromboembolism. Blood 2021, 138 (Suppl. S1), 3023. [Google Scholar] [CrossRef]
  57. Foss, B.; Bruserud, Ø.; Hervig, T. Platelet-Released Supernatants Enhance Hematopoietic Stem Cell Proliferation in Vitro. Platelets 2008, 19, 155–159. [Google Scholar] [CrossRef]
  58. Jungi, T.W.; Spycher, M.O.; Nydegger, U.E.; Barandun, S. Platelet-Leukocyte Interaction: Selective Binding of Thrombin-Stimulated Platelets to Human Monocytes, Polymorphonuclear Leukocytes, and Related Cell Lines. Blood 1986, 67, 629–636. [Google Scholar] [CrossRef]
  59. Moore, K.L.; Stults, N.L.; Diaz, S.; Smith, D.F.; Cummings, R.D.; Varki, A.; McEver, R.P. Identification of a Specific Glycoprotein Ligand for P-Selectin (CD62) on Myeloid Cells. J. Cell Biol. 1992, 118, 445–456. [Google Scholar] [CrossRef] [PubMed]
  60. Ivanov, I.I.; Apta, B.H.R.; Bonna, A.M.; Harper, M.T. Platelet P-Selectin Triggers Rapid Surface Exposure of Tissue Factor in Monocytes. Sci. Rep. 2019, 9, 13397. [Google Scholar] [CrossRef] [PubMed]
  61. Takagi, S.; Tsukamoto, S.; Park, J.; Johnson, K.E.; Kawano, Y.; Moschetta, M.; Liu, C.J.; Mishima, Y.; Kokubun, K.; Manier, S.; et al. Platelets Enhance Multiple Myeloma Progression via Il-1b Upregulation. Clin. Cancer Res. 2018, 24, 2430–2439. [Google Scholar] [CrossRef] [PubMed]
  62. Tsubaki, M.; Komai, M.; Itoh, T.; Imano, M.; Sakamoto, K.; Shimaoka, H.; Ogawa, N.; Mashimo, K.; Fujiwara, D.; Takeda, T.; et al. Inhibition of the Tumour Necrosis Factor-Alpha Autocrine Loop Enhances the Sensitivity of Multiple Myeloma Cells to Anticancer Drugs. Eur. J. Cancer 2013, 49, 3708–3717. [Google Scholar] [CrossRef] [PubMed]
  63. Pignatelli, P.; De Biase, L.; Lenti, L.; Tocci, G.; Brunelli, A.; Cangemi, R.; Riondino, S.; Grego, S.; Volpe, M.; Violi, F. Tumor Necrosis Factor-α as Trigger of Platelet Activation in Patients with Heart Failure. Blood 2005, 106, 1992–1994. [Google Scholar] [CrossRef] [PubMed]
  64. O’Sullivan, L.R.; Meade-Murphy, G.; Gilligan, O.M.; Mykytiv, V.; Young, P.W.; Cahill, M.R. Platelet Hyperactivation in Multiple Myeloma Is Also Evident in Patients with Premalignant Monoclonal Gammopathy of Undetermined Significance. Br. J. Haematol. 2021, 192, 322–332. [Google Scholar] [CrossRef]
  65. Lemancewicz, D.; Bolkun, L.; Mantur, M.; Semeniuk, J.; Kloczko, J.; Dzieciol, J. Bone Marrow Megakaryocytes, Soluble P-Selectin and Thrombopoietic Cytokines in Multiple Myeloma Patients. Platelets 2014, 25, 181–187. [Google Scholar] [CrossRef]
  66. Egan, K.; Cooke, N.; Dunne, E.; Murphy, P.; Quinn, J.; Kenny, D. Platelet Hyporeactivity in Active Myeloma. Thromb. Res. 2014, 134, 747–749. [Google Scholar] [CrossRef]
  67. Kawano, M.; Hirano, T.; Matsuda, T.; Taga, T.; Horii, Y.; Iwato, K.; Asaoku, H.; Tang, B.; Tanabe, O.; Tanaka, H.; et al. Autocrine Generation and Requirement of BSF-2/IL-6 for Human Multiple Myelomas. Nature 1988, 332, 83–85. [Google Scholar] [CrossRef]
  68. Gadó, K.; Domján, G.; Hegyesi, H.; Falus, A. Role of INTERLEUKIN-6 in the Pathogenesis of Multiple Myeloma. Cell Biol. Int. 2000, 24, 195–209. [Google Scholar] [CrossRef]
  69. Steeve, K.T.; Marc, P.; Sandrine, T.; Dominique, H.; Yannick, F. IL-6, RANKL, TNF-Alpha/IL-1: Interrelations in Bone Resorption Pathophysiology. Cytokine Growth Factor Rev. 2004, 15, 49–60. [Google Scholar] [CrossRef] [PubMed]
  70. Kaser, A.; Brandacher, G.; Steurer, W.; Kaser, S.; Offner, F.A.; Zoller, H.; Theurl, I.; Widder, W.; Molnar, C.; Ludwiczek, O.; et al. Interleukin-6 Stimulates Thrombopoiesis through Thrombopoietin: Role in Inflammatory Thrombocytosis. Blood 2001, 98, 2720–2725. [Google Scholar] [CrossRef] [PubMed]
  71. Bilalis, A.; Pouliou, E.; Roussou, M.; Papanikolaou, A.; Tassidou, A.; Economopoulos, T.; Terpos, E. Increased Expression of Platelet Derived Growth Factor Receptor β on Trephine Biopsies Correlates with Advanced Myeloma. J. BUON 2017, 22, 1032–1037. [Google Scholar] [PubMed]
  72. Tsirakis, G.; Pappa, C.A.; Kanellou, P.; Stratinaki, M.A.; Xekalou, A.; Psarakis, F.E.; Sakellaris, G.; Alegakis, A.; Stathopoulos, E.N.; Alexandrakis, M.G. Role of Platelet-Derived Growth Factor-AB in Tumour Growth and Angiogenesis in Relation with Other Angiogenic Cytokines in Multiple Myeloma. Hematol. Oncol. 2012, 30, 131–136. [Google Scholar] [CrossRef] [PubMed]
  73. Tinoco, R.; Otero, D.C.; Takahashi, A.A.; Bradley, L.M. PSGL-1: A New Player in the Immune Checkpoint Landscape. Trends Immunol. 2017, 38, 323. [Google Scholar] [CrossRef] [PubMed]
  74. Atalay, F.; Ateşoğlu, E.B.; Yildiz, S.; Firatli-Tuglular, T.; Karakuş, S.; Bayik, M. Relationship of P-Selectin Glycoprotein Ligand-1 to Prognosis in Patients With Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2015, 15, 164–170. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, Y.; MacLeod, V.; Dai, Y.; Khotskaya-Sample, Y.; Shriver, Z.; Venkataraman, G.; Sasisekharan, R.; Naggi, A.; Torri, G.; Casu, B.; et al. The Syndecan-1 Heparan Sulfate Proteoglycan Is a Viable Target for Myeloma Therapy. Blood 2007, 110, 2041–2048. [Google Scholar] [CrossRef]
  76. Dhodapkar, M.V.; Kelly, T.; Theus, A.; Athota, A.B.; Barlogie, B.; Sanderson, R.D. Elevated Levels of Shed Syndecan-1 Correlate with Tumour Mass and Decreased Matrix Metalloproteinase-9 Activity in the Serum of Patients with Multiple Myeloma. Br. J. Haematol. 1997, 99, 368–371. [Google Scholar] [CrossRef]
  77. Dowling, P.; Hayes, C.; Ting, K.R.; Hameed, A.; Meiller, J.; Mitsiades, C.; Anderson, K.C.; Clynes, M.; Clarke, C.; Richardson, P.; et al. Identification of Proteins Found to Be Significantly Altered When Comparing the Serum Proteome from Multiple Myeloma Patients with Varying Degrees of Bone Disease. BMC Genom. 2014, 15, 904. [Google Scholar] [CrossRef]
  78. Liang, P.; Cheng, S.H.; Cheng, C.K.; Lau, K.M.; Lin, S.Y.; Chow, E.Y.D.; Chan, N.P.H.; Ip, R.K.L.; Wong, R.S.M.; Ng, M.H.L. Platelet Factor 4 Induces Cell Apoptosis by Inhibition of STAT3 via Up-Regulation of SOCS3 Expression in Multiple Myeloma. Haematologica 2013, 98, 288. [Google Scholar] [CrossRef]
  79. Tukijan, F.; Chandrakanthan, M.; Nguyen, L.N. The Signalling Roles of Sphingosine-1-phosphate Derived from Red Blood Cells and Platelets. Br. J. Pharmacol. 2018, 175, 3741. [Google Scholar] [CrossRef] [PubMed]
  80. Li, Q.F.; Wu, C.T.; Guo, Q.; Wang, H.; Wang, L.S. Sphingosine 1-Phosphate Induces Mcl-1 Upregulation and Protects Multiple Myeloma Cells against Apoptosis. Biochem. Biophys. Res. Commun. 2008, 371, 159–162. [Google Scholar] [CrossRef] [PubMed]
  81. Wu, Y.; Liu, Y.; Wang, T.; Jiang, Q.; Xu, F.; Liu, Z. Living Cell for Drug Delivery. Eng. Regen. 2022, 3, 131–148. [Google Scholar] [CrossRef]
  82. Golebiewska, E.M.; Poole, A.W. Platelet Secretion: From Haemostasis to Wound Healing and Beyond. Blood Rev. 2015, 29, 153–162. [Google Scholar] [CrossRef] [PubMed]
  83. Lazar, S.; Goldfinger, L.E. Platelets and Extracellular Vesicles and Their Cross Talk with Cancer. Blood 2021, 137, 3192–3200. [Google Scholar] [CrossRef]
  84. Cohen, S.A.; Trikha, M.; Mascelli, M.A. Potential Future Clinical Applications for the GPIIb/IIIa Antagonist, Abciximab in Thrombosis, Vascular and Oncological Indications. Pathol. Oncol. Res. 2000, 6, 163–174. [Google Scholar] [CrossRef]
  85. Boilard, E. Extracellular Vesicles and Their Content in Bioactive Lipid Mediators: More than a Sack of MicroRNA. J. Lipid Res. 2018, 59, 2037–2046. [Google Scholar] [CrossRef]
  86. Dai, Z.; Zhao, T.; Song, N.; Pan, K.; Yang, Y.; Zhu, X.; Chen, P.; Zhang, J.; Xia, C. Platelets and Platelet Extracellular Vesicles in Drug Delivery Therapy: A Review of the Current Status and Future Prospects. Front. Pharmacol. 2022, 13, 1026386. [Google Scholar] [CrossRef]
  87. Walker, S.; Busatto, S.; Pham, A.; Tian, M.; Suh, A.; Carson, K.; Quintero, A.; Lafrence, M.; Malik, H.; Santana, M.X.; et al. Extracellular Vesicle-Based Drug Delivery Systems for Cancer Treatment. Theranostics 2019, 9, 8001–8017. [Google Scholar] [CrossRef]
  88. McNamee, N.; de la Fuente, L.R.; Santos-Martinez, M.J.; O’Driscoll, L. Proteomics Profiling Identifies Extracellular Vesicles’ Cargo Associated with Tumour Cell Induced Platelet Aggregation. BMC Cancer 2022, 22, 1023. [Google Scholar] [CrossRef]
  89. Xiao, G.; Zhang, Z.; Chen, Q.; Wu, T.; Shi, W.; Gan, L.; Liu, X.; Huang, Y.; Lv, M.; Zhao, Y.; et al. Platelets for Cancer Treatment and Drug Delivery. Clin. Transl. Oncol. 2022, 24, 1231–1237. [Google Scholar] [CrossRef] [PubMed]
  90. Xu, P.; Zuo, H.; Chen, B.; Wang, R.; Ahmed, A.; Hu, Y.; Ouyang, J. Doxorubicin-Loaded Platelets as a Smart Drug Delivery System: An Improved Therapy for Lymphoma OPEN. Sci. Rep. 2017, 7, 42632. [Google Scholar] [CrossRef]
  91. Gay, L.J.; Felding-Habermann, B. Contribution of Platelets to Tumour Metastasis. Nat. Rev. Cancer 2011, 11, 123–134. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, C.; Sun, W.; Ye, Y.; Hu, Q.; Bomba, H.N.; Gu, Z. In Situ Activation of Platelets with Checkpoint Inhibitors for Post-Surgical Cancer Immunotherapy. Nat. Biomed. Eng. 2017, 1, 0011. [Google Scholar] [CrossRef]
  93. Hu, Q.; Sun, W.; Wang, J.; Ruan, H.; Zhang, X.; Ye, Y.; Shen, S.; Wang, C.; Lu, W.; Cheng, K.; et al. Conjugation of Haematopoietic Stem Cells and Platelets Decorated with Anti-PD-1 Antibodies Augments Anti-Leukaemia Efficacy. Nat. Biomed. Eng. 2018, 2, 831. [Google Scholar] [CrossRef]
  94. Li, R.; He, Y.; Zhang, S.; Qin, J.; Wang, J. Cell Membrane-Based Nanoparticles: A New Biomimetic Platform for Tumor Diagnosis and Treatment. Acta Pharm. Sin. B 2018, 8, 14–22. [Google Scholar] [CrossRef]
  95. Hu, Q.; Qian, C.; Sun, W.; Wang, J.; Chen, Z.; Bomba, H.N.; Xin, H.; Shen, Q.; Gu, Z. Engineered Nano-Platelets for Enhanced Treatment of Multiple Myeloma and Thrombus. Adv. Mater. 2016, 28, 9573. [Google Scholar] [CrossRef]
  96. Dai, L.; Liu, Y.; Ding, S.; Wei, X.; Chen, B. Human Nanoplatelets as Living Vehicles for Tumor-Targeted Endocytosis In Vitro and Imaging In Vivo. J. Clin. Med. 2023, 12, 1592. [Google Scholar] [CrossRef]
  97. Kanikarla-Marie, P.; Lam, M.; Menter, D.G.; Kopetz, S. Platelets, Circulating Tumor Cells, and the Circulome. Cancer Metastasis Rev. 2017, 36, 235–248. [Google Scholar] [CrossRef]
  98. Shen, Y.; Lai, Y.; Xu, D.; Xu, L.; Song, L.; Zhou, J.; Song, C.; Wang, J. Diagnosis of Thyroid Neoplasm Using Support Vector Machine Algorithms Based on Platelet RNA-Seq. Endocrine 2021, 72, 758–783. [Google Scholar] [CrossRef]
  99. Sol, N.; in ‘t Veld, S.G.J.G.; Vancura, A.; Tjerkstra, M.; Leurs, C.; Rustenburg, F.; Schellen, P.; Verschueren, H.; Post, E.; Zwaan, K.; et al. Tumor-Educated Platelet RNA for the Detection and (Pseudo)Progression Monitoring of Glioblastoma. Cell Rep. Med. 2020, 1, 100101. [Google Scholar] [CrossRef] [PubMed]
  100. Pastuszak, K.; Supernat, A.; Best, M.G.; In ’t Veld, S.G.J.G.; Łapińska-Szumczyk, S.; Łojkowska, A.; Różański, R.; Żaczek, A.J.; Jassem, J.; Würdinger, T.; et al. ImPlatelet Classifier: Image-Converted RNA Biomarker Profiles Enable Blood-Based Cancer Diagnostics. Mol. Oncol. 2021, 15, 2688–2701. [Google Scholar] [CrossRef] [PubMed]
  101. Xiao, R.; Liu, C.; Zhang, B.; Ma, L. Tumor-Educated Platelets as a Promising Biomarker for Blood-Based Detection of Renal Cell Carcinoma. Front. Oncol. 2022, 12, 689. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Interaction between MM cells and platelets. MM cells express: CD38, S1P, PDGFR, PSGL-1, and Syndecan-1. CD38 could bind to CD31 expressed on resting platelets, whereas PSGL-1 could bind to P-selectin expressed on activated platelets. MM cells release IL-6 and several immunoglobulins, including IgG. IL-6 can indirectly increase platelet count by increasing thrombopoietin production. IgG binds low-affinity Fc receptor (FcR) for the constant region of IgG (FcγRIIa). During activation and subsequent aggregation, platelets release several factors including TGF-β1, PDGF, S1P, and VEGF. PDGF and S1P have affinity for PDGF and S1P receptors, respectively, resulting in angiogenesis and tumor proliferation. Syndecan-1 shed from MM cells can also bind to VEGF, promoting angiogenesis. PSGL-1: P-selectin glycoprotein ligand-1; ADP: adenosine diphosphate; IgG: immunoglobulin G; IL-6: interleukin-6; PAR: protease-activated receptor; TGF-β1: transforming growth factor β1; PDGF: platelet-derived growth factor; PDGFR: platelet-derived growth factor receptor.
Figure 1. Interaction between MM cells and platelets. MM cells express: CD38, S1P, PDGFR, PSGL-1, and Syndecan-1. CD38 could bind to CD31 expressed on resting platelets, whereas PSGL-1 could bind to P-selectin expressed on activated platelets. MM cells release IL-6 and several immunoglobulins, including IgG. IL-6 can indirectly increase platelet count by increasing thrombopoietin production. IgG binds low-affinity Fc receptor (FcR) for the constant region of IgG (FcγRIIa). During activation and subsequent aggregation, platelets release several factors including TGF-β1, PDGF, S1P, and VEGF. PDGF and S1P have affinity for PDGF and S1P receptors, respectively, resulting in angiogenesis and tumor proliferation. Syndecan-1 shed from MM cells can also bind to VEGF, promoting angiogenesis. PSGL-1: P-selectin glycoprotein ligand-1; ADP: adenosine diphosphate; IgG: immunoglobulin G; IL-6: interleukin-6; PAR: protease-activated receptor; TGF-β1: transforming growth factor β1; PDGF: platelet-derived growth factor; PDGFR: platelet-derived growth factor receptor.
Ijms 24 15855 g001
Table 1. Revised International Staging System (R-ISS).
Table 1. Revised International Staging System (R-ISS).
R-ISS StageCriteria
ISβ2M 1<3.5 mg/L
Serum albumin≥3.5 g/dL
Chromosomal abnormalities by iFISH 2Standard risk
LDH 3Normal (<upper normal limit)
IINot R-ISS stage I or III
IIISβ2M
and either
Chromosomal abnormalities by FISH
or
LDH
≥5.5 mg/L

*High risk


High (>upper normal limit)
1 Sβ2M—serum β2 microglobulin; 2 FISH—fluorescence in situ hybridization; 3 LDH—lactate dehydrogenase. Taken from [10]. *High risk is defined as presence of del(17p) and/or translocation t(4;14) and/or translocation t(14;1.6)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kulkarni, A.; Bazou, D.; Santos-Martinez, M.J. Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems. Int. J. Mol. Sci. 2023, 24, 15855. https://doi.org/10.3390/ijms242115855

AMA Style

Kulkarni A, Bazou D, Santos-Martinez MJ. Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems. International Journal of Molecular Sciences. 2023; 24(21):15855. https://doi.org/10.3390/ijms242115855

Chicago/Turabian Style

Kulkarni, Anushka, Despina Bazou, and Maria José Santos-Martinez. 2023. "Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems" International Journal of Molecular Sciences 24, no. 21: 15855. https://doi.org/10.3390/ijms242115855

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop