Liquid biopsies have been considered the holy grail in achieving effective cancer management, with blood tests offering a minimally invasive, safe, and sensitive alternative or complementary approach for tissue biopsies. Currently, blood-based liquid biopsy measurements focus on the evaluation of biomarker types, including circulating tumor DNA, circulating tumor cells, extracellular vesicles (exosomes and oncosomes), and tumor-educated platelets (TEPs). Despite the potential of individual techniques, each has its own advantages and disadvantages. Here, we provide further insight into TEPs.

Liquid biopsies could provide a potential revolution in cancer diagnostics as a minimally invasive method for detecting and monitoring cancer. Liquid biopsies may provide an accurate and comprehensive snapshot of the tumor and its microenvironment on multiple levels.1  Liquid biopsies enable early detection (screening), as well as prognosis for the individual patient for tumor stage and identification of new targets for personalized treatment. In addition, blood tests can be employed for pretreatment classification for personalized therapy, early-therapy response monitoring, and “real-time” assessment of treatment effectiveness, as well as follow-up and early detection of disease recurrence. Multiple blood-based biosources are currently evaluated (reviewed in Heitzer et al,2  Babayan and Pantel,3  Aravanis et al,4  Best et al,5  and Wan et al6 ), including TEPs. Platelets, originating as anucleate cells from megakaryocytes, have emerged as central players in the systemic and local responses to tumor growth.5,7,8  In the past few years, methods for isolation and analysis of spliced TEP messenger RNA (mRNA) were developed to detect cancer with high accuracy.9-13  The development of TEPs as a potential liquid biopsy biosource, however, is a consequence of many studies on the role of platelets in cancer over the past few centuries (Figure 1).

Figure 1.

Historical timeline of platelets in relation to cancer. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 1.

Historical timeline of platelets in relation to cancer. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Platelets are known for their function in coagulation of blood and wound healing, but their relationship with cancer has been studied extensively as well (reviewed in Franco et al,14  Lambert et al,15  Menter et al,16  Naderi-Meshkin and Ahmadiankia,17  Haemmerle et al,18  Olsson et al,19  Xu et al,20  Zhang et al,21  and Mancuso and Santagostino22 ). Two observations fundamental to the development of the concept of TEPs were made in the 19th century. First, in 1868, Trousseau shared his observation that spontaneous coagulation is common in cancerous patients,23  indicating that circulating platelets are affected by cancer.24  Second, in 1877, Billroth described “thrombi filled with specific tumor elements” as part of tumor metastasis,25  indicating a direct interaction between tumor cells and platelets.26-28  Evolving technology subsequently elucidated parts of the complex interactions among megakaryocytes, platelets, and cancer, leading to the concept of TEPs.5,8,11,29,30 

In 1906, Wright reported that platelets are detached portions of the cytoplasm of megakaryocytes, which were identified in the bone marrow and spleen.31  However, circulating megakaryocytes were also observed in certain pathologic conditions,32  including cancer.33  In 2010, Zaslavsky et al showed that the number of megakaryocytes in the bone marrow increases in response to cancer.34  Kerr et al showed that communication between tumor and bone is mediated by platelets via tumor-derived proteins stored in granules.35  The increase in megakaryocytes and platelets was considered to be a systemic response to suppress tumor growth, established by elevated thrombospondin-1 levels in platelets.34  The relationship between platelet numbers and cancer was identified in the 1960s.36,37  A large-scale platelet screening study among 14 000 individuals revealed that thrombocytosis is associated with a variety of diseases, of which neoplasms in different organs resembled the largest group.36  In 1968, Gasic and colleagues showed a correlation between thrombocytopenia and cancer metastasis in mice. Injection with platelet-rich plasma reversed the antimetastatic effect observed for thrombocytopenic mice.38  In a follow-up study, they reported that in some instances, tumor cell–induced thrombocytopenia is accompanied by accumulation of platelets in the lung and that platelets with aggregating capacities usually correlate with lung metastases.37  It was recently demonstrated that the lung also is a site of platelet biogenesis,39  altogether suggesting a potential function of the lung (besides bone marrow) in the interaction between platelets and cancer metastases. The exact function of platelets from different origins has not been investigated yet. However, since ∼700 differentially expressed genes between lung and bone marrow megakaryocytes were identified,39  it is not unlikely that the RNA of platelets originating in bone marrow and lung differs as well. Hence, it may be of interest to study the spliced RNA profiles of the platelet populations derived from bone marrow and lung in response to various tumor-associated signals. It was shown that platelets also interact directly with intravasating and circulating tumor cells (CTCs), thereby altering platelet signaling.40-46  In 2017, Jiang et al were able to isolate platelet-covered CTCs from blood and observed that CTCs were extensively covered by platelets.45  Besides platelet numbers, the proteome and size of platelets and the platelet/immune cell ratio47-49  have also been reported to change in the presence of cancer.50-52  Moreover, meta-analyses showed that platelet inhibitors such as aspirin may be associated with a reduced risk of several cancer types.53  Experiments showed that inhibition of platelets by aspirin can affect their ability to induce cancer cell proliferation.54,55  Further research is warranted to determine how platelet inhibitors can be optimally employed as anticancer agents.18,20 

For decades, it was believed that the platelet content was static, because platelets are cell fragments lacking a nucleus, and therefore no transcription and translation was expected. However, Warshaw et al performed experiments suggesting the ability of protein synthesis in platelets,56  a process extensively reviewed by Weyrich and colleagues.57  Booyse et al confirmed protein synthesis by platelets and suggested that the life span of the platelet mRNA determines the life span of the platelets.58  Subsequently, it was confirmed that platelet protein synthesis requires ribosomes.59  It took over 20 years until the involvement of mRNA in protein synthesis was confirmed by Newman et al using polymerase chain reaction.60 

Power et al investigated the role of platelets in inflammation and used a complementary DNA library of human platelets and polymerase chain reaction to show the presence of mRNA coding for several chemokines in platelets.61  Weyrich et al demonstrated that protein synthesis is part of a signal-dependent activation response in human platelets,62  showing thrombin-induced 4E-BP1 phosphorylation and Bcl-3 expression in platelets. Also intra-platelet signaling by protein-tyrosine phosphorylation was revealed,63  and Nadal-Wollbold et al showed that thrombin-induced ERK2 activation is activated by conventional protein kinase Cs independently of Raf-1 and B-Raf activation.64  External stimuli, including lipopolysaccharide,65  P-selectin ligands,66  and thrombin,63,64  activate kinase pathway signaling, processes possibly resulting in “platelet education” that may lead to transformation of naive platelets into protumorigenic platelets.67  More explicit, Kuznetsov and colleagues found that platelets that absorb tumor-derived cytokines aid vessel formation at tumor sites.67 

Various stimuli can alter platelet signaling and induce protein synthesis. The mechanism behind such observations was investigated by Denis et al in 2005. They showed that platelets possess a functional spliceosome including small nuclear RNAs, splicing proteins, and endogenous pre-mRNAs. The spliceosome can be induced to cause pre-mRNA splicing in response to stimuli.68  Schwertz et al reported that splicing of intron-rich tissue factor pre-mRNA into mature mRNA in healthy donors is controlled by Clk1.69  The involvement of RNA-binding proteins in the splicing mechanism of platelets was suggested as well.5,12  Platelets contain many different RNA species,70-73  including microRNAs72,74,75  and circular RNAs.76  It was shown that microRNA modulation induces proteome reorganization as well.77  A study combining proteomics and transcriptomics of platelets identified ∼8000 transcripts of which a set would undergo intron removal during platelet activation, concluding that maturation of specific pre-mRNAs contributes to the dynamic fine-tuning of the transcriptome.78  Thus, platelets arise as anucleate cytoplasmic fragments from megakaryocytes and retain megakaryocyte-derived cytoplasmic pre-mRNA, of which at least some are spliced into mature mRNAs and translated into proteins in response to external stimuli.79,80  Hence, tumor cells can directly stimulate platelet protein synthesis and platelet signaling. However, the protein content of platelets can consist of megakaryocyte-derived proteins, endocytosed proteins, and proteins translated in individual platelets. Therefore, it is important to determine which proteins are truly tumor derived and which proteins may fluctuate due to tumor-independent processes. Besides education via direct platelet–tumor cell interaction, tumor-associated biomolecules such as proteins35,81  and RNA9,10,82-84  can be sequestered by platelets. Examples of directly transferred transcripts include the cancer RNA biomarkers EGFRvIII,9,83  PCA3,9  EML4-ALK,82  KRAS, EGFR, and PIK3CA mutants,10  FOLH1, KLK2, KLK3, and NPY.84  Similar results have been reported for endothelial RNA transcripts taken up by platelets.85  The combination of specific splice events in response to external signals and the capacity of platelets to directly ingest (spliced) circulating mRNA provides TEPs with a highly dynamic transcriptome, with potential applicability as liquid biopsies for cancer diagnostics.9 

Interestingly, platelets can respond to external stimuli by releasing RNA signaling complexes to other cells86,87  via microparticles, suggesting an intricate communicational crosstalk network. The role of platelet-derived microparticles (PMPs) in cancer development and progression has been previously reviewed.87  PMPs are the most abundant vesicle population in peripheral blood, accounting for ∼70% to 90% of all extracellular vesicles. It was found that PMPs are formed after platelet activation and are involved in angiogenesis, metastasis and multidrug resistance.87  Conversely, tumor-derived vesicles have the capability to sequester in platelets proteins and nucleic acids, possibly also including circulating free DNA, which seems to be inherent to platelets during their entire life span.5,9,88  Further research is needed to determine if platelets have a selection mechanism for specific vesicle sizes and types, and which internalization mechanisms are used.

Calverley et al and Nilsson et al demonstrated in 2010 and 2011, respectively, that the RNA profiles of platelets of cancer patients are altered as compared with healthy donors.9,89  In 2015, Best et al performed extensive RNA sequencing to determine differentially spliced RNA profiles in platelets from cancer patients (comprising 6 different types of cancer) and healthy individuals.10  This resulted in a predictive “pan-cancer” test with 96% accuracy.10  In addition, in a multiclass test across the 6 different tumor types (non-small cell lung carcinoma [NSCLC], colorectal cancer, glioblastoma, pancreatic cancer, hepatobiliary cancer, and breast cancer), the location of the primary tumor was correctly identified with 71% accuracy, indicating significant tumor-type-specific splicing within platelets. Detecting cancer using spliced TEP profiles was confirmed in a large follow-up study on platelets of NSCLC patients,12  and confirmed by others.90-92  Various individual spliced transcripts were detected in a validation study of NSCLC patients; the most significantly increased transcripts were CFL1, ACOT7, and ARPC1B, whereas DDX5, RPS5, and EEF1B2 were decreased, independent of age, smoking status, and blood storage, as well as various inflammatory conditions.12  Although no direct experiments were performed, it is likely that the spliced RNA detected in platelets from NSCLC patients is derived mostly from megakaryocytes and spliced in response to tumor-associated signals rather than tumor derived and ingested by the platelets. In addition, gene ontology analysis demonstrated that the spliced TEP transcripts correlated to translation, RNA-binding proteins, and intraplatelet signaling in low transcript splicing–levels NSCLC samples and interplatelet signaling and immune response in high transcript splicing–level samples. Additional clustering analyses revealed correlations between specific immune signaling pathways in TEPs of NSCLC patients and platelet homeostasis in platelets of individuals not diagnosed with cancer.12  Although further research is warranted, the mechanisms by which altered spliced RNA profiles may arise in TEPs include tumor-induced alteration of RNA transferred by megakaryocytes into platelets,34  signal-dependent platelet RNA splicing,68,69,78  exon skipping and alternative splicing, differential RNA-binding protein activity,12  and altered platelet aging/turnover.5,12 

Platelet activation is associated with the initiation of coagulation cascades. During this process, platelets change shape and release the content of their granules, leading to platelet aggregation.93  A study comparing platelets from patients with ovarian cancer and those with benign ovarian tumors described that platelets are not hyperactive in patients with ovarian cancer.94  Cho and colleagues found that platelets increase the proliferation rate of ovarian cancer. Of note, they observed that direct contact between platelets and cancer cells or an intact structure of platelets was not required for the proliferative response. Moreover, blocking platelet adhesive surface proteins (GPIa, GPIIbIIIa, and P-selectin) did not diminish the proliferative effect of platelets. Aspirin only partially inhibited the proliferative effect of platelets on ovarian cancer cells.95  Although platelet activation can be induced by tumor cells, Best et al showed the absence of significant platelet activation during blood collection and storage of the analyzed platelet samples.12  Therefore, it is unlikely that the observed TEP RNA profiles are entirely driven by platelet activation. However, further exploration of the effects of both platelet activation and anticoagulation on TEP profiles is desired. Studies aiming at improving the power of TEP classifiers could include comparisons of changes in spliced RNA profiles in procoagulant states vs cancer-derived platelet activation, which would allow us to understand changes in platelet function induced by cancer vs other prothrombotic events. Clearly, further work is needed to validate these findings and evaluate potential events that generate noise or affect the diagnostic detection levels. Recently, a standardized protocol for thromboSeq, as well as TEP classifier development software, has been made available13  to further evaluate the diagnostic power of platelets. Studies comparing and combining different techniques are required to evaluate the (dis)advantages of individual biosources and the potential of combining them into a next-generation multianalyte liquid biopsy test.

The development and implementation of liquid biopsies in clinical settings requires a strong interdisciplinary effort, with a wide range of scientific competencies. Key will be to identify and select the best TEP biomarkers using the most sensitive techniques, including ultra-deep, massive parallel, and long-read sequencing of TEP transcripts (perhaps also including detection of epigenetic and epitranscriptomic features). Combining liquid biopsy sources for the detection and localization of cancer has proven to be effective.96  TEPs may offer certain advantages over other blood-based biosources, including their abundance and easy isolation, high-quality RNA, and capacity to process RNA in response to external signals.5,11,30  Combinatorial analysis of TEPs with complementary biosources such as extracellular vesicles, circulating tumor DNA, and CTCs, but possibly also imaging and protein markers, warrants consideration as next-generation biomarker troves, thereby seeking optimal diagnostic synergy.

Financial support was provided by the European Research Council (grants H2020-MSCA 765492, 713727, and 336540), the Netherlands Organisation for Scientific Research (grant 91711366), and the Dutch Cancer Society (grant VU2015-8080).

Contribution: S.G.J.G.I.t.V. and T.W. wrote the manuscript.

Conflict-of-interest disclosure: T.W. received funding from Illumina Inc. and is a shareholder of GRAIL Inc. S.G.J.G.I.t.V. declares no competing financial interests.

Correspondence: Thomas Wurdinger, Amsterdam UMC, VU University Medical Center, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; e-mail: t.wurdinger@vumc.nl.

1.
Bardelli
A
,
Pantel
K
.
Liquid biopsies: what we do not know (yet)
.
Cancer Cell
.
2017
;
31
(
2
):
172
-
179
.
2.
Heitzer
E
,
Haque
IS
,
Roberts
CES
,
Speicher
MR
.
Current and future perspectives of liquid biopsies in genomics-driven oncology
.
Nat Rev Genet
.
2019
;
20
(
2
):
71
-
88
.
3.
Babayan
A
,
Pantel
K
.
Advances in liquid biopsy approaches for early detection and monitoring of cancer
.
Genome Med
.
2018
;
10
(
1
):
21
.
4.
Aravanis
AM
,
Lee
M
,
Klausner
RD
.
Next-generation sequencing of circulating tumor dna for early cancer detection
.
Cell
.
2017
;
168
(
4
):
571
-
574
.
5.
Best
MG
,
Wesseling
P
,
Wurdinger
T
.
Tumor-educated platelets as a noninvasive biomarker source for cancer detection and progression monitoring
.
Cancer Res
.
2018
;
78
(
13
):
3407
-
3412
.
6.
Wan
JCM
,
Massie
C
,
Garcia-Corbacho
J
, et al
.
Liquid biopsies come of age: towards implementation of circulating tumour DNA
.
Nat Rev Cancer
.
2017
;
17
(
4
):
223
-
238
.
7.
Plantureux
L
,
Crescence
L
,
Dignat-George
F
,
Panicot-Dubois
L
,
Dubois
C
.
Effects of platelets on cancer progression
.
Thromb Res
.
2018
;
164
(
suppl 1
):
S40
-
S47
.
8.
McAllister
SS
,
Weinberg
RA
.
The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis
.
Nat Cell Biol
.
2014
;
16
(
8
):
717
-
727
.
9.
Nilsson
RJA
,
Balaj
L
,
Hulleman
E
, et al
.
Blood platelets contain tumor-derived RNA biomarkers
.
Blood
.
2011
;
118
(
13
):
3680
-
3683
.
10.
Best
MGG
,
Sol
N
,
Kooi
I
, et al
.
RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics
.
Cancer Cell
.
2015
;
28
(
5
):
666
-
676
.
11.
Best
MG
,
Vancura
A
,
Wurdinger
T
.
Platelet RNA as a circulating biomarker trove for cancer diagnostics
.
J Thromb Haemost
.
2017
;
15
(
7
):
1295
-
1306
.
12.
Best
MG
,
Sol
N
,
In ‘t Veld
SGJG
, et al
.
Swarm intelligence-enhanced detection of non-small-cell lung cancer using tumor-educated platelets
.
Cancer Cell
.
2017
;
32
(
2
):
238
-
252.e9
.
13.
Best
MG
,
In ‘t Veld
SGJG
,
Sol
N
,
Wurdinger
T
.
RNA sequencing and swarm intelligence–enhanced classification algorithm development for blood-based disease diagnostics using spliced blood platelet RNA
.
Nat Protoc
.
In press
.
14.
Franco
AT
,
Corken
A
,
Ware
J
.
Platelets at the interface of thrombosis, inflammation, and cancer
.
Blood
.
2015
;
126
(
5
):
582
-
588
.
15.
Lambert
AW
,
Pattabiraman
DR
,
Weinberg
RA
.
Emerging biological principles of metastasis
.
Cell
.
2017
;
168
(
4
):
670
-
691
.
16.
Menter
DG
,
Kopetz
S
,
Hawk
E
, et al
.
Platelet “first responders” in wound response, cancer, and metastasis
.
Cancer Metastasis Rev
.
2017
;
36
(
2
):
199
-
213
.
17.
Naderi-Meshkin
H
,
Ahmadiankia
N
.
Cancer metastasis versus stem cell homing: Role of platelets
.
J Cell Physiol
.
2018
;
233
(
12
):
9167
-
9178
.
18.
Haemmerle
M
,
Stone
RL
,
Menter
DG
,
Afshar-Kharghan
V
,
Sood
AK
.
The Platelet Lifeline to Cancer: Challenges and Opportunities
.
Cancer Cell
.
2018
;
33
(
6
):
965
-
983
.
19.
Olsson
AK
,
Cedervall
J
.
The pro-inflammatory role of platelets in cancer
.
Platelets
.
2018
;
29
(
6
):
569
-
573
.
20.
Xu
XR
,
Yousef
GM
,
Ni
H
.
Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents
.
Blood
.
2018
;
131
(
16
):
1777
-
1789
.
21.
Zhang
Q
,
Liu
H
,
Zhu
Q
, et al
.
Patterns and functional implications of platelets upon tumor “education”
.
Int J Biochem Cell Biol
.
2017
;
90
:
68
-
80
.
22.
Mancuso
ME
,
Santagostino
E
.
Platelets: much more than bricks in a breached wall
.
Br J Haematol
.
2017
;
178
(
2
):
209
-
219
.
23.
Trousseau
A
.
Lectures on Clinical Medicine, Delivered at the Hotel-Dieu, Paris. Translated from Editions of 1868 by J.R. Cormack
.
London
:
The New Sydenham Society
;
1868
.
24.
Hisada
Y
,
Mackman
N
.
Cancer-associated pathways and biomarkers of venous thrombosis
.
Blood
.
2017
;
130
(
13
):
1499
-
1506
.
25.
Billroth
T
.
Lectures on Surgical Pathology and Therapeutics: a Handbook for Students and Practitioners
.
London
:
The New Sydenham Society
;
1877
.
26.
Kanikarla-Marie
P
,
Lam
M
,
Menter
DG
,
Kopetz
S
.
Platelets, circulating tumor cells, and the circulome
.
Cancer Metastasis Rev
.
2017
;
36
(
2
):
235
-
248
.
27.
Contursi
A
,
Sacco
A
,
Grande
R
,
Dovizio
M
,
Patrignani
P
.
Platelets as crucial partners for tumor metastasis: from mechanistic aspects to pharmacological targeting
.
Cell Mol Life Sci
.
2017
;
74
(
19
):
3491
-
3507
.
28.
Leblanc
R
,
Peyruchaud
O
.
Metastasis: new functional implications of platelets and megakaryocytes
.
Blood
.
2016
;
128
(
1
):
24
-
31
.
29.
Tjon-Kon-Fat
L-A
,
Sol
N
,
Wurdinger
T
,
Nilsson
RJA
.
Platelet RNA in cancer diagnostics
.
Semin Thromb Hemost
.
2018
;
44
(
2
):
135
-
141
.
30.
Sol
N
,
Wurdinger
T
.
Platelet RNA signatures for the detection of cancer
.
Cancer Metastasis Rev
.
2017
;
36
(
2
):
263
-
272
.
31.
WRIGHT JH
.
The origin and nature of the blood plates
.
Boston Med Surg J
.
1906
;
154
(
23
):
643
-
645
.
32.
Minot
GR
.
Megacaryocytes in the peripheral circulation
.
J Exp Med
.
1922
;
36
(
1
):
1
-
7
.
33.
Hume
R
,
West
JT
,
Malmgren
RA
,
Chu
EA
.
Quantitative observations of circulating megakaryocytes in the blood of patients with cancer
.
N Engl J Med
.
1964
;
270
(
3
):
111
-
117
.
34.
Zaslavsky
A
,
Baek
K-H
,
Lynch
RC
, et al
.
Platelet-derived thrombospondin-1 is a critical negative regulator and potential biomarker of angiogenesis
.
Blood
.
2010
;
115
(
22
):
4605
-
4613
.
35.
Kerr
BA
,
McCabe
NP
,
Feng
W
,
Byzova
TV
.
Platelets govern pre-metastatic tumor communication to bone
.
Oncogene
.
2013
;
32
(
36
):
4319
-
4324
.
36.
Levin
J
,
Conley
CL
.
Thrombocytosis associated with malignant disease
.
Arch Intern Med
.
1964
;
114
(
4
):
497
-
500
.
37.
Gasic
GJ
,
Gasic
TB
,
Galanti
N
,
Johnson
T
,
Murphy
S
.
Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease
.
Int J Cancer
.
1973
;
11
(
3
):
704
-
718
.
38.
Gasic
GJ
,
Gasic
TB
,
Stewart
CC
.
Antimetastatic effects associated with platelet reduction
.
Proc Natl Acad Sci USA
.
1968
;
61
(
1
):
46
-
52
.
39.
Lefrançais
E
,
Ortiz-Muñoz
G
,
Caudrillier
A
, et al
.
The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors
.
Nature
.
2017
;
544
(
7648
):
105
-
109
.
40.
Micalizzi
DS
,
Maheswaran
S
,
Haber
DA
.
A conduit to metastasis: circulating tumor cell biology
.
Genes Dev
.
2017
;
31
(
18
):
1827
-
1840
.
41.
Labelle
M
,
Hynes
RO
.
The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination
.
Cancer Discov
.
2012
;
2
(
12
):
1091
-
1099
.
42.
Reymond
N
,
d’Água
BB
,
Ridley
AJ
.
Crossing the endothelial barrier during metastasis
.
Nat Rev Cancer
.
2013
;
13
(
12
):
858
-
870
.
43.
Labelle
M
,
Begum
S
,
Hynes
RO
.
Platelets guide the formation of early metastatic niches
.
Proc Natl Acad Sci USA
.
2014
;
111
(
30
):
E3053
-
E3061
.
44.
Labelle
M
,
Begum
S
,
Hynes
RO
.
Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis
.
Cancer Cell
.
2011
;
20
(
5
):
576
-
590
.
45.
Jiang
X
,
Wong
KHK
,
Khankhel
AH
, et al
.
Microfluidic isolation of platelet-covered circulating tumor cells
.
Lab Chip
.
2017
;
17
(
20
):
3498
-
3503
.
46.
Schlesinger
M
.
Role of platelets and platelet receptors in cancer metastasis
.
J Hematol Oncol
.
2018
;
11
(
1
):
125
.
47.
Ma
JY
,
Ke
LC
,
Liu
Q
.
The pretreatment platelet-to-lymphocyte ratio predicts clinical outcomes in patients with cervical cancer: a meta-analysis
.
Medicine (Baltimore)
.
2018
;
97
(
43
):
e12897
.
48.
Zhang
LX
,
Wei
ZJ
,
Xu
AM
,
Zang
JH
.
Can the neutrophil-lymphocyte ratio and platelet-lymphocyte ratio be beneficial in predicting lymph node metastasis and promising prognostic markers of gastric cancer patients? Tumor maker retrospective study
.
Int J Surg
.
2018
;
56
:
320
-
327
.
49.
Temur
I
,
Kucukgoz Gulec
U
,
Paydas
S
,
Guzel
AB
,
Sucu
M
,
Vardar
MA
.
Prognostic value of pre-operative neutrophil/lymphocyte ratio, monocyte count, mean platelet volume, and platelet/lymphocyte ratio in endometrial cancer
.
Eur J Obstet Gynecol Reprod Biol
.
2018
;
226
:
25
-
29
.
50.
Sabrkhany
S
,
Kuijpers
MJE
,
Griffioen
AW
,
Oude Egbrink
MGA
.
Platelets: the holy grail in cancer blood biomarker research?
Angiogenesis
.
2019
;
22
(
1
):
1
-
2
.
51.
Sabrkhany
S
,
Kuijpers
MJE
,
Knol
JC
, et al
.
Exploration of the platelet proteome in patients with early-stage cancer
.
J Proteomics
.
2018
;
177
:
65
-
74
.
52.
Sabrkhany
S
,
Kuijpers
MJE
,
van Kuijk
SMJ
, et al
.
A combination of platelet features allows detection of early-stage cancer
.
Eur J Cancer
.
2017
;
80
:
5
-
13
.
53.
Qiao
Y
,
Yang
T
,
Gan
Y
,
Li
W
,
Wang
C
,
Gong
Y
,
Lu
Z
.
Associations between aspirin use and the risk of cancers: a meta-analysis of observational studies
.
BMC Cancer
.
2018
;
18
(
1
):
288
.
54.
Mitrugno
A
,
Sylman
JL
,
Rigg
RA
, et al
.
Carpe low-dose aspirin: the new anti-cancer face of an old anti-platelet drug
.
Platelets
.
2018
;
29
(
8
):
773
-
778
.
55.
Patrignani
P
,
Patrono
C
.
Aspirin, platelet inhibition and cancer prevention
.
Platelets
.
2018
;
29
(
8
):
779
-
785
.
56.
Warshaw
AL
,
Laster
L
,
Shulman
NR
.
The stimulation by thrombin of glucose oxidation in human platelets
.
J Clin Invest
.
1966
;
45
(
12
):
1923
-
1934
.
57.
Weyrich
AS
,
Schwertz
H
,
Kraiss
LW
,
Zimmerman
GA
.
Protein synthesis by platelets: historical and new perspectives
.
J Thromb Haemost
.
2009
;
7
(
2
):
241
-
246
.
58.
Booyse
FM
,
Rafelson
ME
Jr
.
Stable messenger RNA in the synthesis of contractile protein in human platelets
.
Biochim Biophys Acta
.
1967
;
145
(
1
):
188
-
190
.
59.
Booyse
FM
,
Rafelson
ME
Jr
.
Studies on human platelets. I. synthesis of platelet protein in a cell-free system
.
Biochim Biophys Acta
.
1968
;
166
(
3
):
689
-
697
.
60.
Newman
PJ
,
Gorski
J
,
White
GC
II
,
Gidwitz
S
,
Cretney
CJ
,
Aster
RH
.
Enzymatic amplification of platelet-specific messenger RNA using the polymerase chain reaction
.
J Clin Invest
.
1988
;
82
(
2
):
739
-
743
.
61.
Power
CA
,
Clemetson
JM
,
Clemetson
KJ
,
Wells
TNC
.
Chemokine and chemokine receptor mRNA expression in human platelets
.
Cytokine
.
1995
;
7
(
6
):
479
-
482
.
62.
Weyrich
AS
,
Dixon
DA
,
Pabla
R
, et al
.
Signal-dependent translation of a regulatory protein, Bcl-3, in activated human platelets
.
Proc Natl Acad Sci USA
.
1998
;
95
(
10
):
5556
-
5561
.
63.
Ezumi
Y
,
Takayama
H
,
Okuma
M
.
Thrombopoietin, c-Mpl ligand, induces tyrosine phosphorylation of Tyk2, JAK2, and STAT3, and enhances agonists-induced aggregation in platelets in vitro
.
FEBS Lett
.
1995
;
374
(
1
):
48
-
52
.
64.
Nadal-Wollbold
F
,
Pawlowski
M
,
Lévy-Toledano
S
,
Berrou
E
,
Rosa
JP
,
Bryckaert
M
.
Platelet ERK2 activation by thrombin is dependent on calcium and conventional protein kinases C but not Raf-1 or B-Raf
.
FEBS Lett
.
2002
;
531
(
3
):
475
-
482
.
65.
Brown
GT
,
McIntyre
TM
.
Lipopolysaccharide signaling without a nucleus: kinase cascades stimulate platelet shedding of proinflammatory IL-1β-rich microparticles
.
J Immunol
.
2011
;
186
(
9
):
5489
-
5496
.
66.
Crovello
CS
,
Furie
BC
,
Furie
B
.
Rapid phosphorylation and selective dephosphorylation of P-selectin accompanies platelet activation
.
J Biol Chem
.
1993
;
268
(
20
):
14590
-
14593
.
67.
Kuznetsov
HS
,
Marsh
T
,
Markens
BA
, et al
.
Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells
.
Cancer Discov
.
2012
;
2
(
12
):
1150
-
1165
.
68.
Denis
MM
,
Tolley
ND
,
Bunting
M
, et al
.
Escaping the nuclear confines: signal-dependent pre-mRNA splicing in anucleate platelets
.
Cell
.
2005
;
122
(
3
):
379
-
391
.
69.
Schwertz
H
,
Tolley
ND
,
Foulks
JM
, et al
.
Signal-dependent splicing of tissue factor pre-mRNA modulates the thrombogenicity of human platelets
.
J Exp Med
.
2006
;
203
(
11
):
2433
-
2440
.
70.
Bray
PF
,
McKenzie
SE
,
Edelstein
LC
, et al
.
The complex transcriptional landscape of the anucleate human platelet
.
BMC Genomics
.
2013
;
14
(
1
):
1
.
71.
Rowley
JW
,
Chappaz
S
,
Corduan
A
, et al
.
Dicer1-mediated miRNA processing shapes the mRNA profile and function of murine platelets
.
Blood
.
2016
;
127
(
14
):
1743
-
1751
.
72.
Nagalla
S
,
Shaw
C
,
Kong
X
, et al
.
Platelet microRNA-mRNA coexpression profiles correlate with platelet reactivity
.
Blood
.
2011
;
117
(
19
):
5189
-
5197
.
73.
Rowley
JW
,
Oler
AJ
,
Tolley
ND
, et al
.
Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes
.
Blood
.
2011
;
118
(
14
):
e101
-
e111
.
74.
Boilard
E
,
Belleannée
C
.
(Dicer)phering roles of microRNA in platelets
.
Blood
.
2016
;
127
(
14
):
1733
-
1734
.
75.
Schubert
S
,
Weyrich
AS
,
Rowley
JW
.
A tour through the transcriptional landscape of platelets
.
Blood
.
2014
;
124
(
4
):
493
-
502
.
76.
Alhasan
AA
,
Izuogu
OG
,
Al-Balool
HH
, et al
.
Circular RNA enrichment in platelets is a signature of transcriptome degradation
.
Blood
.
2016
;
127
(
9
):
e1
-
e11
.
77.
Cimmino
G
,
Tarallo
R
,
Nassa
G
, et al
.
Activating stimuli induce platelet microRNA modulation and proteome reorganisation
.
Thromb Haemost
.
2015
;
114
(
1
):
96
-
108
.
78.
Nassa
G
,
Giurato
G
,
Cimmino
G
, et al
.
Splicing of platelet resident pre-mRNAs upon activation by physiological stimuli results in functionally relevant proteome modifications
.
Sci Rep
.
2018
;
8
(
1
):
498
.
79.
Bahou
WF
,
Gnatenko
DV
.
Platelet transcriptome: the application of microarray analysis to platelets
.
Semin Thromb Hemost
.
2004
;
30
(
4
):
473
-
484
.
80.
Qiu
Y
,
Ciciliano
J
,
Myers
DR
,
Tran
R
,
Lam
WA
.
Platelets and physics: How platelets “feel” and respond to their mechanical microenvironment
.
Blood Rev
.
2015
;
29
(
6
):
377
-
386
.
81.
Klement
GL
,
Yip
T-T
,
Cassiola
F
, et al
.
Platelets actively sequester angiogenesis regulators
.
Blood
.
2009
;
113
(
12
):
2835
-
2842
.
82.
Nilsson
RJA
,
Karachaliou
N
,
Berenguer
J
, et al
.
Rearranged EML4-ALK fusion transcripts sequester in circulating blood platelets and enable blood-based crizotinib response monitoring in non-small-cell lung cancer
.
Oncotarget
.
2016
;
7
(
1
):
1066
-
1075
.
83.
Luo
C-L
,
Xu
Z-G
,
Chen
H
, et al
.
LncRNAs and EGFRvIII sequestered in TEPs enable blood-based NSCLC diagnosis
.
Cancer Manag Res
.
2018
;
10
:
1449
-
1459
.
84.
Tjon-Kon-Fat
L-A
,
Lundholm
M
,
Schröder
M
, et al
.
Platelets harbor prostate cancer biomarkers and the ability to predict therapeutic response to abiraterone in castration resistant patients
.
Prostate
.
2018
;
78
(
1
):
48
-
53
.
85.
Clancy
L
,
Beaulieu
LM
,
Tanriverdi
K
,
Freedman
JE
.
The role of RNA uptake in platelet heterogeneity
.
Thromb Haemost
.
2017
;
117
(
5
):
948
-
961
.
86.
Laffont
B
,
Corduan
A
,
Plé
H
, et al
.
Activated platelets can deliver mRNA regulatory Ago2•microRNA complexes to endothelial cells via microparticles
.
Blood
.
2013
;
122
(
2
):
253
-
261
.
87.
Żmigrodzka
M
,
Guzera
M
,
Miśkiewicz
A
,
Jagielski
D
,
Winnicka
A
.
The biology of extracellular vesicles with focus on platelet microparticles and their role in cancer development and progression
.
Tumour Biol
.
2016
;
37
(
11
):
14391
-
14401
.
88.
Gomes
FG
,
Sandim
V
,
Almeida
VH
, et al
.
Breast-cancer extracellular vesicles induce platelet activation and aggregation by tissue factor-independent and -dependent mechanisms
.
Thromb Res
.
2017
;
159
:
24
-
32
.
89.
Calverley
DC
,
Phang
TL
,
Choudhury
QG
, et al
.
Significant downregulation of platelet gene expression in metastatic lung cancer
.
Clin Transl Sci
.
2010
;
3
(
5
):
227
-
232
.
90.
Li
D
,
Yang
W
,
Zhang
Y
, et al
.
Genomic analyses based on pulmonary adenocarcinoma in situ reveal early lung cancer signature
.
BMC Med Genomics
.
2018
;
11
(
S5 Suppl 5
):
106
.
91.
Sheng
M
,
Dong
Z
,
Xie
Y
.
Identification of tumor-educated platelet biomarkers of non-small-cell lung cancer
.
OncoTargets Ther
.
2018
;
11
:
8143
-
8151
.
92.
Xue
L
,
Xie
L
,
Song
X
,
Song
X
.
Expression and significance of ACIN1 mRNA in platelets of lung cancer [in Chinese]
.
Zhongguo Fei Ai Za Zhi
.
2018
;
21
(
9
):
677
-
681
.
93.
Yun
S-H
,
Sim
E-H
,
Goh
R-Y
,
Park
J-I
,
Han
J-Y
.
Platelet activation: the mechanisms and potential biomarkers
.
BioMed Res Int
.
2016
;
2016
:
9060143
.
94.
Feng
S
,
Kroll
MH
,
Nick
AM
,
Sood
AK
,
Afshar-Kharghan
V
.
Platelets are not hyperreactive in patients with ovarian cancer
.
Platelets
.
2016
;
27
(
7
):
716
-
718
.
95.
Cho
MS
,
Bottsford-Miller
J
,
Vasquez
HG
, et al
.
Platelets increase the proliferation of ovarian cancer cells
.
Blood
.
2012
;
120
(
24
):
4869
-
4872
.
96.
Cohen
JD
,
Li
L
,
Wang
Y
, et al
.
Detection and localization of surgically resectable cancers with a multi-analyte blood test
.
Science
.
2018
;
359
(
6378
):
926
-
930
.
Sign in via your Institution