References
- Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele
J, Vardiman JW. WHO Classification of Tumours of Haematopoietic and
Lymphoid Tissues. 4th ed. Lyon, France: IARC Press; 2008
- Jaffe E.S.et al. Classification of lymphoid neoplasms: the microscope
as a tool for disease discovery. Blood. 2008; 112(12):4384–99. doi:
10.1182/blood-2008-07-077982,
- Laurini, J. A., Perry, A. M., Boilesen, E., Diebold, J., MacLennan, K.
A., Müller-Hermelink, H. K., Nathwani, B. N., Armitage, J. O., &
Weisenburger, D. D.(2012). Classification of non-Hodgkin lymphoma in
Central and South America: a review of 1028 cases. Blood, 120(24),
4795-4801. Accessed January 17, 2019. https://doi.org/10.1)
- Teras, Lauren R., et al. ”2016 US lymphoid malignancy statistics by
World Health Organization subtypes.” CA: a cancer journal for
clinicians 66.6 (2016): 443-459
- Morton, L. M., Wang, S. S., Devesa, S. S., Hartge, P., Weisenburger,
D. D., & Linet, M. S. (2006). Lymphoma incidence patterns by WHO
subtype in the United States, 1992-2001. Blood, 107(1), 265-276.
Accessed January 17, 2019.
https://doi.org/10.1182/blood-2005-06-2508
- Shenoy, Pareen J., et al. ”Racial differences in the presentation and
outcomes of diffuse large B‐cell lymphoma in the United States.”
Cancer 117.11 (2011): 2530-2540
- Howlader N, Noon AM, Krapcho M, et al., eds. SEER Cancer Statistics
Review, 1975-2013, National Cancer Institute, Bethesda, MD, based on
November 2015 SEER data submission, posted to the SEER web site, April
2016. https://seer.cancer.gov/csr/1975_2013/
- Sant M, Minicozzi P, Mounier M, et al; EUROCARE-5 Working Group.
Survival for haematological malignancies in Europe between 1997 and
2008 by region and age: results of EUROCARE-5, a population-based
study. Lancet Oncol. 2014;15(9):931-942
- Crump, M., Neelapu, S. S., Farooq, U., Van Den Neste, E., Kuruvilla,
J., Westin, J., Gisselbrecht, C. (2017). Outcomes in refractory
diffuse large B-cell lymphoma: results from the international
SCHOLAR-1 study. Blood, 130(16), 1800–1808.
doi:10.1182/blood-2017-03-769620
- Lenz, G., Rutherford, S. C., Davies, A., Zinzani, P. L., Salles, G.,
Hasskarl, J., Margunato-Debay, S., Rodrigues, F., Nientker, L.,
Balradj, J., & Alleman, C. The Burden of Relapsed/Refractory (R/R)
Diffuse Large B-Cell Lymphoma (DLBCL): A Systematic Literature Review
(SLR). Blood, 132(Suppl 1), 2241. Accessed January 17, 2019.
https://doi.org/10.1182/blood-2018-99-112878.)
- Friedberg, J.W. (2011) Relapsed/refractory diffuse large B‐cell
lymphoma. ASH Education Program Book, 2011, 498–505.)
- Vose, J. M., Weisenburger, D. D., Loberiza, F. R., Arevalo, A., Bast,
M., Armitage, J., … & Armitage, J. O. (2010). Late relapse in
patients with diffuse large B‐cell lymphoma. British journal of
haematology, 151(4), 354-358
- Broséus, J., Chen, G., Hergalant, S., Ramstein, G., Mounier, N.,
Guéant, J. L., … Houlgatte, R. Relapsed diffuse large B-cell
lymphoma present different genomic profiles between early and late
relapses. Oncotarget, 7(51), 83987–84002.
doi:10.18632/oncotarget.9793
- Gisselbrecht C, Glass B, Mounier N, et al. Salvage regimens with
autologous transplantation for relapsed large B-cell lymphoma in the
rituximab era. J Clin Oncol. 2010;28(27):4184-4190, 10
- R. Vaidya, T. E. Witzig, Prognostic factors for diffuse large B-cell
lymphoma in the R(X)CHOP era, Annals of Oncology, Volume 25, Issue 11,
November 2014, Pages 2124–2133,
https://doi.org/10.1093/annonc/mdu109
- Biccler, Jorne L., Sandra Eloranta, Peter de Nully Brown, Henrik
Frederiksen, Mats Jerkeman, Judit Jørgensen, Lasse Hjort Jakobsen,
Karin E. Smedby, Martin Bøgsted, and Tarec C. El-Galaly. ”Optimizing
Outcome Prediction in Diffuse Large B-Cell Lymphoma by Use of Machine
Learning and Nationwide Lymphoma Registries: A Nordic Lymphoma Group
Study.” JCO clinical cancer informatics 2 (2018): 1-13
- International Non-Hodgkin’s Lymphoma Prognostic Factors Project.
(1993). A predictive model for aggressive non-Hodgkin’s lymphoma. New
England Journal of Medicine, 329(14), 987-994
- Biccler, Jorne Lionel, Tarec Christoffer El-Galaly, Martin Bøgsted,
Judit Jørgensen, Peter de Nully Brown, Christian Bjørn Poulsen, Jørn
Starklint et al. ”Clinical prognostic scores are poor predictors of
overall survival in various types of malignant
lymphomas.” Leukemia & lymphoma 60, no. 6 (2019): 1580-1583.
- Sehn, L. H., Berry, B., Chhanabhai, M., Fitzgerald, C., Gill, K.,
Hoskins, P., … & Gascoyne, R. D. (2007). The revised International
Prognostic Index (R-IPI) is a better predictor of outcome than the
standard IPI for patients with diffuse large B-cell lymphoma treated
with R-CHOP. Blood, 109(5), 1857-1861
- Alizadeh, A. A., Eisen, M. B., Davis, R. E., Ma, C., Lossos, I. S.,
Rosenwald, A., … & Powell, J. I. (2000). Distinct types of diffuse
large B-cell lymphoma identified by gene expression profiling. Nature,
403(6769), 503
- Christof Schneider, Laura Pasqualucci, Riccardo Dalla-Favera,
Molecular pathogenesis of diffuse large B-cell lymphoma, Seminars in
Diagnostic Pathology Volume 28, Issue 2,2011,Pages 167-177
- Rosenwald A, Wright G, Chan WC, et al. The use of molecular profiling
to predict survival after chemotherapy for diffuse large-B-cell
lymphoma. N Engl J Med. 2002;346:1937–1947.
- Vitolo, Umberto, et al. ”Obinutuzumab or rituximab plus
cyclophosphamide, doxorubicin, vincristine, and prednisone in
previously untreated diffuse large B-cell lymphoma.” Journal of
Clinical Oncology 35.31 (2017): 3529-3537
- Scott DW, Mottok A, Ennishi D, et al. Prognostic Significance of
Diffuse Large B-Cell Lymphoma Cell of Origin Determined by Digital
Gene Expression in Formalin-Fixed Paraffin-Embedded Tissue Biopsies. J
Clin Oncol. 2015;33(26):2848–2856. doi:10.1200/JCO.2014.60.2383
- Xu-Monette, Zijun Y. et al ”Mutational profile and prognostic
significance of TP53 in diffuse large B-cell lymphoma patients treated
with R-CHOP: report from an International DLBCL Rituximab-CHOP
Consortium Program Study.” Blood 120.19 (2012): 3986-3996. Web. 01
April. 2019.
- Wilson, W. H., Young, R. M., Schmitz, R., Yang, Y., Pittaluga, S.,
Wright, G., … & De Vos, S. (2015). Targeting B cell receptor
signaling with ibrutinib in diffuse large B cell
lymphoma. Nature medicine , 21 (8), 922
- Scott, D. W., Wright, G. W., Williams, P. M., Lih, C., Walsh,
W., Jaffe, E. S., Rosenwald, A., Campo, E., Chan, W. C., Connors, J.
M., Smeland, E. B., Mottok, A., Braziel, R. M., Ott, G., Delabie,
J., Tubbs, R. R., Cook, J. R., Weisenburger, D. D., Greiner, T.
C., Glinsmann-Gibson, B. J., Fu, K., Staudt, L. M., Gascoyne, R. D.,
& Rimsza, L. M. (2014). Determining cell-of-origin subtypes of
diffuse large B-cell lymphoma using gene expression in formalin-fixed
paraffin-embedded tissue. Blood, 123(8), 1214-1217. Accessed
July 07, 2019. https://doi.org/10.1182/blood-2013-11-536433.)
- Hans, C. P., Weisenburger, D. D., Greiner, T. C., Gascoyne, R. D.,
Delabie, J., Ott, G., Müller-Hermelink, H. K., Campo, E., Braziel, R.
M., Jaffe, E. S., Pan, Z., Farinha, P., Smith, L. M., Falini, B.,
Banham, A. H., Rosenwald, A., Staudt, L. M., Connors, J. M., Armitage,
J. O., & Chan, W. C. (2004). Confirmation of the molecular
classification of diffuse large B-cell lymphoma by
immunohistochemistry using a tissue microarray. Blood, 103(1),
275-282.Accessed April 10, 2019.
https://doi.org/10.1182/blood-2003-05-1545
- Visco C, Li Y, Xu-Monette ZY, et al. Comprehensive gene expression
profiling and immunohistochemical studies support application of
immunophenotypic algorithm for molecular subtype classification in
diffuse large B-cell lymphoma: a report from the International DLBCL
Rituximab-CHOP Consortium Program Study [published correction
appears in Leukemia. 2014 Apr;28(4):980]. Leukemia.
2012;26(9):2103–2113. doi:10.1038/leu.2012.83
- William W.L. Choi, Dennis D. Weisenburger, Timothy C. Greiner, Miguel
A. Piris, Alison H. Banham, Jan Delabie, Rita M. Braziel, Huimin Geng,
Javeed Iqbal, Georg Lenz, Julie M. Vose, Christine P. Hans, Kai Fu,
Lynette M. Smith, Min Li, Zhongfeng Liu, Randy D. Gascoyne, Andreas
Rosenwald, German Ott, Lisa M. Rimsza, Elias Campo, Elaine S. Jaffe,
David L. Jaye, Louis M. Staudt and Wing C. Chan. A New Immunostain
Algorithm Classifies Diffuse Large B-Cell Lymphoma into Molecular
Subtypes with High Accuracy. Clin Cancer Res September 1 2009 (15)
(17) 5494-5502; DOI: 10.1158/1078-0432.CCR-09-0113
- Meyer PN, Fu K, Greiner TC, et al. Immunohistochemical methods for
predicting cell of origin and survival in patients with diffuse large
B-cell lymphoma treated with rituximab. J Clin Oncol.
2010;29(2):200–207. doi:10.1200/JCO.2010.30.036
- Gutiérrez-García, G., Cardesa-Salzmann, T., Climent, F.,
González-Barca, E., Mercadal, S., Mate, J. L., Sancho, J. M.,
Arenillas, L., Serrano, S., Escoda, L., Martínez, S., Valera, A.,
Martínez, A., Jares, P., Pinyol, M., García-Herrera, A.,
Martínez-Trillos, A., Giné, E., Villamor, N., Campo, E., Colomo, L.,
López-Guillermo, A., & , . (2011) Blood, 117(18), 4836-4843. Accessed
January 26, 2019. https://doi.org/10.1182/blood-2010-12-322362).
- Yoon, Nara, et al. ”Cell-of-origin of diffuse large B-cell lymphomas
determined by the Lymph2Cx assay: better prognostic indicator than
Hans algorithm.” Oncotarget 8.13 (2017): 22014,
- 25. Nyman H, Adde M, Karjalainen-Lindsberg ML, Taskinen M, Berglund M,
Amini RM, Blomqvist C, Enblad G, Leppa S. Prognostic impact of
immunohistochemically defined germinal center phenotype in diffuse
large B-cell lymphoma patients treated with immunochemotherapy. Blood.
2007; 109:4930–4935.
- Gleeson M, Hawkes EA, Cunningham D, Jack A, Linch D. Caution in the
Use of Immunohistochemistry for Determination of Cell of Origin in
Diffuse Large B-Cell Lymphoma. J Clin Oncol. 2015; 33:3215–3216
- Schmitz, Roland, et al. ”Genetics and pathogenesis of diffuse large
B-cell lymphoma.” New England Journal of Medicine378.15 (2018):
1396-1407
- Chapuy, Bjoern, et al. ”Molecular subtypes of diffuse large B cell
lymphoma are associated with distinct pathogenic mechanisms and
outcomes.” Nature medicine 24.5 (2018): 679
- Lopez, Juanita S., and Udai Banerji. ”Combine and conquer: challenges
for targeted therapy combinations in early phase trials.” Nature
reviews Clinical oncology 14.1 (2017): 57
- Wierda, William G., et al. ”NCCN guidelines insights: chronic
lymphocytic leukemia/small lymphocytic lymphoma, version 1.2017.”
Journal of the National Comprehensive Cancer Network 15.3 (2017):
293-31
- B.D. Cheson, R.I. Fisher, S.F. Barrington, et al.Recommendations for
initial evaluation, staging, and response assessment of Hodgkin and
non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol, 32
(2014), pp. 3059-3068
- Han HS, Escalon MP, Hsiao B, et al. High incidence of false‐positive
PET scans in patients with aggressive non‐Hodgkin’s lymphoma treated
with rituximab‐containing regimens. Ann Oncol 2009;20:309–318.
- 36. El‐Galaly T, Prakash V, Christiansen I, et al. Efficacy of routine
surveillance with positron emission tomography/computed tomography in
aggressive non‐Hodgkin lymphoma in complete remission: status in a
single center. Leuk Lymphoma2011;52:597–60
- Guppy AE, Tebbutt NC, Norman A, . The role of surveillance CT scans in
patients with diffuse large B-cell non-Hodgkin’s lymphoma. Leuk
Lymphoma 2003;44:123–125
- Cohen, J. B., Behera, M., Thompson, C. A., & Flowers, C. R. (2017).
Evaluating surveillance imaging for diffuse large B-cell lymphoma and
Hodgkin lymphoma. Blood, 129(5), 561-564. Accessed February 02,
2019.https://doi.org/10.1182/blood-2016-08-685073
- Durani, U., Asante, D., Heien, H. C., Thompson, C. A., Halfdanarson,
T. R., Peethambaram, P., Quevedo, J. F., Villasboas, J. C., & Go, R.
S. (2018). Changes in Imaging Surveillance of Diffuse Large B-Cell
Lymphoma Survivors after Publication of the American Society of
Hematology Choosing Wisely®Recommendations. Blood, 132(Suppl 1), 618.
Accessed February 04,
2019.https://doi.org/10.1182/blood-2018-99-119179
- Smith-Bindman R, Lipson J, Marcus R. Radiation dose associated with
common computed tomography examinations and the associated lifetime
attributable risk of cancer. Arch Intern Med 2009; 169:2078–2086
- Alix-Panabières C, Pantel K. Circulating tumor cells: liquid biopsy of
cancer. Clin Chem. 2013 Jan; 59(1):110-8
- Krebs, M. G. et al. Molecular analysis of circulating tumour cells —
biology and biomarkers. Nat. Rev. Clin. Oncol. 11, 129–144 (2014
- Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid
biopsies into the management of cancer. Nat Rev Clin Oncol. 2017
Sep;14(9):531-548. doi: 10.1038/nrclinonc.2017.14. Epub 2017 Mar 2
- Haber, D. A. & Velculescu, V. E. Blood-based analyses of cancer:
circulating tumor cells and circulating tumor DNA. Cancer Discov. 4,
650–661 (2014
- Riethdorf S, Fritsche H, Muller V, Rau T, Schindlbeck C, Rack B, et
al. Detection of circulating tumor cells in peripheral blood of
patients with metastatic breast cancer:a validation study of the
CellSearch system. Clin Cancer Res. 2007;13:920–8
- Talasaz AH, Powell AA, Huber DE, Berbee JG, Roh KH, Yu W, et al.
Isolating highly enriched populations of circulating epithelial cells
and other rare cells from blood using a magnetic sweeper device. Proc
Natl Acad Sci U S A. 2009;106:3970–5
- Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et
al. Isolation of rare circulating tumour cells in cancer patients by
microchip technology. Nature. 2007;450:1235–9
- Brock, G., Castellanos-Rizaldos, E., Hu, L., Coticchia, C., & Skog,
J. (2015). Liquid biopsy for cancer screening, patient stratification
and monitoring. Translational Cancer Research, 4(3), 280-290
- Bettegowda, C. et al. Detection of circulating tumor DNA in early- and
late-stage human malignancies.Sci. Transl Med. 6, 224ra24 (2014
- Thierry, A. R. et al. Clinical validation of the detection of KRAS and
BRAF mutations from circulating tumor DNA. Nat. Med. 20, 430–435
(2014
- Narayan, A. et al. Ultrasensitive measurement of hotspot mutations in
tumor DNA in blood using error- suppressed multiplexed deep
sequencing. Cancer Res. 72, 3492–3498 (2012),
- Liggett, T. et al. Differential methylation of cell-free circulating
DNA among patients with pancreatic cancer versus chronic pancreatitis.
Cancer 116, 1674–1680 (2010),
- Sturgeon, S. R. et al. Detection of promoter methylation of tumor
suppressor genes in serum DNA of breast cancer cases and benign breast
disease controls. Epigenetics 7, 1258–1267 (2012)
- Cristiano, Stephen, Alessandro Leal, Jillian Phallen, Jacob Fiksel,
Vilmos Adleff, Daniel C. Bruhm, Sarah Østrup Jensen et al.
”Genome-wide cell-free DNA fragmentation in patients with
cancer.” Nature (2019): 1.
- Roschewski M, Dunleavy K, Pittaluga S, et al. Circulating
tumour DNA and CT monitoring in 124patients with untreated
diffuse large B-cell lymphoma: a correlative biomarker study. Lancet
125Oncol. 2015;16(5):541-549.1264.
- Sebastián, E., Alcoceba, M., Balanzategui, A., Marín, L.,
Montes-Moreno, S., Flores, T., … & Corral, R. (2012). Molecular
characterization of immunoglobulin gene rearrangements in diffuse
large B-cell lymphoma: antigen-driven origin and IGHV4-34 as a
particular subgroup of the non-GCB subtype. The American journal
of pathology , 181 (5), 1879-1888.
- Mitterbauer-Hohendanner, G., Mannhalter, C., Winkler, K., Mitterbauer,
M., Skrabs, C., Chott, A., … & Jaeger, U. (2004). Prognostic
significance of molecular staging by PCR-amplification of
immunoglobulin gene rearrangements in diffuse large B-cell lymphoma
(DLBCL). Leukemia , 18 (6), 1102.
- Hurley JH, Odorizzi G. Get on the exosome bus with ALIX. Nat Cell
Biol. 2012; 14:654-655. https://doi.org/10.1038/ncb2530
- Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small
particle, big player. J. Hematol. Oncol. 8, 83 (2015)
- Février B, Raposo G. Exosomes: endosomal-derived vesicles shipping
extracellular messages. Curr Opin Cell Biol 2004;16(4):415-421
- Mathieu, M., Martin-Jaular, L., Lavieu, G., & Théry, C. (2019).
Specificities of secretion and uptake of exosomes and other
extracellular vesicles for cell-to-cell communication. Nature cell
biology, 21(1), 9.,
- C. Lässer, M. Eldh, J. Lötvall Isolation and characterization of
RNA-containing exosomes J. Vis. Exp., 59 (2012), pp. 3037-3047,
- Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO.
Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism
of genetic exchange between cells. Nat. Cell Biol. 9(6), 654–659
(2007), C.
- Subra, K. Laulagnier, B. Perret, M. Record. Exosome lipidomics
unravels lipid sorting at the level of multivesicular bodies
Biochimie, 89 (2007), pp. 205-212,
- B.S. Batista, W.S. Eng, K.T. Pilobello, K.D. Hendricks-Muñoz, L.K.
Mahal Identification of a conserved glycan signature for
microvesicles. J. Proteome Res., 10 (2011), pp. 4624-4633
- Crescitelli, Rossella, Cecilia Lässer, Tamas G. Szabó, Agnes Kittel,
Maria Eldh, Irma Dianzani, Edit I. Buzás, and Jan Lötvall. ”Distinct
RNA profiles in subpopulations of extracellular vesicles: apoptotic
bodies, microvesicles and exosomes.” Journal of extracellular vesicles
2, no. 1 (2013): 20677
- Jeffrey S. Schorey and Sanchita Bhatnagar1. Exosome Function: From
Tumor Immunology to Pathogen Biology. Traffic 2008;
9:871–881Blackwell Munksgaard
- Xia, Bing, et al. ”The central roles of exosomes in hematological
malignancies: A new frontier review.” Bioscience Biotechnology
Research Comminications11.3 (2018): 347-355
- Mikhail A. Livshits, Elena Khomyakova, Evgeniy G. Evtushenko, Vassili
N. Lazarev, Nikolay A. Kulemin, Svetlana E. Semina, Edward V.
Generozov & Vadim M. Govorun. Isolation of exosomes by differential
centrifugation: Theoretical analysis of a commonly used protocol
Scientific Reports volume 5, Article number: 17319 (2015
- Momen Heravi F, Balaj L, Alian S, Trachtenberg A, Hochberg F, Skog J,
et al. Impact of biofluid viscosity on size and sedimentation
efficiency of the isolated microvesicles. Front Physiol. 2012;3:162
- Lane, R. E., Korbie, D., Anderson, W., Vaidyanathan, R. & Trau, M.
Analysis of exosome purification methods using a model liposome system
and tunable-resistive pulse sensing. Sci Rep. 5, 7639 (2015)
- Witwer, K. W. et al. Standardization of sample collection, isolation
and analysis methods in extracellular vesicle research. J Extracell
Vesicles. 2, 20360 (2013)
- Théry, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and
Characterization of Exosomes From Cell Culture Supernatants and
Biological Fluids. Current Protocols in Cell Biology: Hoboken, NJ,
2006; Chapter 3, Unit 3.22, DOI: 10.1002/0471143030.cb0322s30,
- Vidal, M.; Mangeat, P.; Hoekstra, D. Aggregation Reroutes Molecules
From a Recycling to a Vesicle-Mediated Secretion Pathway During
Reticulocyte Maturation J. Cell Sci. 1997, 110, 1867– 1877
- Huilin Shao, Hyungsoon Im, Cesar M. Castro, Xandra Breakefield, Ralph
Weissleder, and Hakho Lee. New Technologies for Analysis of
Extracellular Vesicles. Chemical Reviews 2018 118 (4), 1917-1950
DOI: 10.1021/acs.chemrev.7b00534
- Heinemann, M. L., Ilmer, M., Silva, L. P., Hawke, D. H., Recio, A.,
Vorontsova, M. A., … & Vykoukal, J. (2014). Benchtop isolation and
characterization of functional exosomes by sequential filtration.
Journal of Chromatography A, 1371, 125-135.
- Alvarez, M.L.; Khosroheidari, M.; Kanchi Ravi, R.; DiStefano, J.K.
Comparison of protein, microRNA, and mRNA yields using different
methods of urinary exosome isolation for the discovery of kidney
disease biomarkers. Kidney Int. 2012, 82, 1024–1032
- Gámez-Valero, A., Monguió-Tortajada, M., Carreras-Planella, L., Beyer,
K., & Borràs, F. E. (2016). Size-Exclusion Chromatography-based
isolation minimally alters Extracellular Vesicles’ characteristics
compared to precipitating agents. Scientific reports, 6, 33641.
- Taylor D, Zacharias W, Gercel Taylor C. Exosome isolation for
proteomic analyses and RNA profiling. Methods Mol Biol. 2011;
728:235-46
- Taylor, D. D., & Shah, S. (2015). Methods of isolating extracellular
vesicles impact down-stream analyses of their cargoes. Methods, 87,
3-10). The disadvantage of this method is significant co-isolation
with non-exosomal molecules and limited suitability of recovered
exosomes for downstream analyses.
- B. J. Tauro, D. W. Greening, R. A. Mathias et al., “Comparison of
ultracentrifugation, density gradient separation, and immunoaffinity
capture methods for isolating human colon cancer cell line
LIM1863-derived exosomes,” Methods, vol. 56, no. 2, pp. 293–304,
2012.,
- K. A. Konadu, M. B. Huang, W. Roth et al., “Isolation of exosomes
from the plasma of HIV-1 positive individuals,” Journal of Visualized
Experiments, vol. 2016, no. 107, Article ID e53495, 2016
- Zeringer, E., Barta, T., Li, M., & Vlassov, A. V. (2015). Strategies
for isolation of exosomes. Cold Spring Harbor Protocols, 2015(4),
pdb-top074476). Immunoaffinity methods yield isolates of high purity
but have low capacity.
- Witwer, K. W., Buzas, E. I., Bemis, L. T., Bora, A., Lässer, C., &
Lötvall, J. (2013). Nolte-’t Hoen EN, Piper MG, Sivaraman S, Skog J,
Théry C, Wauben MH, Hochberg F. Standardization of sample collection,
isolation and analysis methods in extracellular vesicle research. J
Extracell Vesicles, 2(10.3402
- Lee, K.; Shao, H.; Weissleder, R.; Lee, H. Acoustic Purification of
Extracellular Microvesicles ACS Nano 2015, 9, 2321– 2327 DOI:
10.1021/nn506538f
- Liu, C., Guo, J., Tian, F., Yang, N., Yan, F., Ding, Y., … & Sun,
J. (2017). Field-free isolation of exosomes from extracellular
vesicles by microfluidic viscoelastic flows. ACS nano, 11(7),
6968-6976
- R. C. Lai, R. W. Y. Yeo, K. H. Tan, and S. K. Lim, “Exosomes for drug
delivery—a novel application for the mesenchymal stem cell,”
Biotechnology Advances, vol. 31, no. 5, pp. 543–551, 2013
- Vlassov, A. V., Magdaleno, S., Setterquist, R., & Conrad, R. (2012).
Exosomes: current knowledge of their composition, biological
functions, and diagnostic and therapeutic potentials. Biochimica et
Biophysica Acta (BBA)-General Subjects, 1820(7), 940-948
- Alimirzaie, Sahar, Maryam Bagherzadeh, and Mohammad R. Akbari. ”Liquid
Biopsy in Breast Cancer: A Comprehensive Review.” Clinical genetics
(2019
- Rutherford, Sarah C., Angela Fachel, Sheng Li, Yanwen Jiang, Maria
Pilar Dominguez, Doron Betel, and Rita Shaknovich. ”DLBCL-Derived
Exosomes Provide Key Insights into Genomic Landscape in Cell of Origin
and May Lead to a Novel Method of Surveillance and Therapeutic
Intervention.” (2017): 5132-5132.
- Feng, Yuhua, Meizuo Zhong, Shan Zeng, Leyuan Wang, Ping Liu, Xiangyu
Xiao, and Yiping Liu. ”Exosome-derived miRNAs as predictive biomarkers
for diffuse large B-cell lymphoma chemotherapy resistance.”
Epigenomics 11, no. 1 (2018): 35-51
- Nasrin Zare, Shaghayegh Haghjooy Javanmard, Valiollah Mehrzad, Nahid
Eskandari & Amirhosein Kefayat (2019) Evaluation of exosomal miR-155,
let-7g and let-7i levels as a potential noninvasive biomarker among
refractory/relapsed patients, responsive patients and patients
receiving R-CHOP, Leukemia & Lymphoma, DOI:
10.1080/10428194.2018.1563692
- Di, C., Jiang, Y., Li, M., Juan, X., & Xu, C. (2018). Circulating
Exosomal microRNA Signature As a Noninvasive Biomarker for Diagnosis
of Diffuse Large B-Cell Lymphoma. Blood, 132(Suppl 1), 5406. Accessed
March 10, 2019. https://doi.org/10.1182/blood-2018-99-115940
- Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, Montabana E, Lang
UE, Fu Q, Fong L, Blelloch R. Suppression of Exosomal PD-L1 Induces
Systemic Anti-Tumor Immunity and Memory. Cell. 2019 Apr
4;177(2):414-427.e13. doi: 10.1016/j.cell.2019.02.016
- G Raposo, H W Nijman, W Stoorvogel, R Liejendekker, C V Harding, C J
Melief, H J Geuze. B lymphocytes secrete antigen-presenting vesicles.
Journal of Experimental Medicine Mar 1996, 183 (3) 1161-1172; DOI:
10.1084/jem.183.3.1161
- 61. Chaput, Nathalie, and Clotilde Théry. ”Exosomes: immune properties
and potential clinical implementations.” In Seminars in
immunopathology, vol. 33, no. 5, pp. 419-440. Springer-Verlag, 2011.
- Chen, Zhenzhen, Liangshun You, Lei Wang, Xianbo Huang, Hui Liu, Ju
ying Wei, Li Zhu, and Wenbin Qian. ”Dual effect of DLBCL-derived EXOs
in lymphoma to improve DC vaccine efficacy in vitro while favor
tumorgenesis in vivo.” Journal of Experimental & Clinical Cancer
Research 37, no. 1 (2018): 190
- Koch, Raphael, et al. ”Populational equilibrium through
exosome-mediated Wnt signaling in tumor progression of diffuse large
B-cell lymphoma.” Blood 123.14 (2014): 2189-2198.
- Koch, R., Aung, T., Vogel, D., Chapuy, B., Wenzel, D., Becker, S., …
& Truemper, L. (2016). Nuclear trapping through inhibition of
exosomal export by indomethacin increases cytostatic efficacy of
doxorubicin and pixantrone. Clinical cancer research, 22(2), 395-404
- Aung, Thiha, et al. ”Exosomal evasion of humoral immunotherapy in
aggressive B-cell lymphoma modulated by ATP-binding cassette
transporter A3.” Proceedings of the National Academy of Sciences
108.37 (2011): 15336-15341
- J Castillo, V Bernard, F A San Lucas, K Allenson, M Capello, D U Kim,
P Gascoyne, F C Mulu, B M Stephens, J Huang, H Wang, A A Momin, R O
Jacamo, M Katz, R Wolff, M Javle, G Varadhachary, I I Wistuba, S
Hanash, A Maitra, H Alvarez, Surfaceome profiling enables isolation of
cancer-specific exosomal cargo in liquid biopsies from pancreatic
cancer patients, Annals of Oncology, Volume 29, Issue 1, January 2018,
Pages 223–229, https://doi.org/10.1093/annonc/mdx542
- Mizutani, Kosuke, Riyako Terazawa, Koji Kameyama, Taku Kato, Kengo
Horie, Tomohiro Tsuchiya, Kensaku Seike et al. ”Isolation of prostate
cancer-related exosomes.” Anticancer research 34, no. 7 (2014):
3419-3423.
- Jeppesen, Dennis K., Aidan M. Fenix, Jeffrey L. Franklin, James N.
Higginbotham, Qin Zhang, Lisa J. Zimmerman, Daniel C. Liebler et al.
”Reassessment of exosome composition.” Cell 177, no. 2 (2019): 428-445