1
|
Hanafy RM, Demian SR, Abou-Shamaa LA, Ghallab O, Osman EM. In-vitro Modulation of mTOR-HIF-1α Axis by TLR7/8 Agonist (Resiquimod) in B-Chronic Lymphocytic Leukemia. Indian J Hematol Blood Transfus 2023; 39:537-545. [PMID: 37786827 PMCID: PMC10542076 DOI: 10.1007/s12288-023-01649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/20/2023] [Indexed: 10/04/2023] Open
Abstract
Targeting toll-like receptors (TLRs), via TLR agonists, has been implicated in the regulation of immunometabolism. B-chronic lymphocytic leukemia (B-CLL) represents a suitable model for B-cell derived malignancies with shifted metabolic adaptations. Several signaling pathways have been found to be critical in metabolic reprogramming of CLL, including mechanistic target of rapamycin- hypoxia inducible factor-1α (mTOR- HIF-1α) pathway, the main metabolic regulator of glycolysis. Here, we investigated the effect of TLR7/8 agonist (Resiquimod) on the expression of mTOR and HIF-1α in patients with CLL. B cells were purified using Rosettesep Human B cell Enrichment Cocktail (Stem cell Technologies, Vancouver, BC, Canada#15,024) from peripheral venous blood of CLL patients (n = 20) and healthy individuals (n = 15). Isolated B cells were then cultured in both presence and absence of Resiquimod. Gene expression of mTOR and HIF-1α were assessed using qRT-PCR. Resiquimod significantly decreased mTOR and HIF-1α gene expression in both CLL (p < 0.001and p < 0.001, respectively) and Normal B cells (p = 0.004 and p = 0.001, respectively). Resiquimod may reprogram immunometabolism of malignant B-CLL cells via down-regulation of key glycolytic metabolic actors, mTOR and HIF-1α genes. Accordingly, Resiquimod may be an adjuvant as a therapeutic tool for CLL, which needs to be studied further. Supplementary Information The online version contains supplementary material available at 10.1007/s12288-023-01649-y.
Collapse
Affiliation(s)
- Rana M. Hanafy
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Soheir R. Demian
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Lobna A. Abou-Shamaa
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - O. Ghallab
- Internal Medicine Department (Hematology Unit), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman M. Osman
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Zhou Y, Dong Z, Andarge H, Li W, Pappas D. Nanoparticle modification of microfluidic cell separation for cancer cell detection and isolation. Analyst 2020; 145:257-267. [DOI: 10.1039/c9an01719d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We present a nanoparticle surface modification approach to improve the microfluidic performance in detecting cancer cells. Multiple cancer cell lines were included in this work, and the capture ability of the chip with surface modification reached a significant increase.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| | - Ziye Dong
- Department of Chemical Engineering
- Texas Tech University
- Lubbock
- USA
| | - Hermella Andarge
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| | - Wei Li
- Department of Chemical Engineering
- Texas Tech University
- Lubbock
- USA
| | - Dimitri Pappas
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| |
Collapse
|
3
|
Corrupted coordination of epigenetic modifications leads to diverging chromatin states and transcriptional heterogeneity in CLL. Nat Commun 2019; 10:1874. [PMID: 31015400 PMCID: PMC6478836 DOI: 10.1038/s41467-019-09645-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/22/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer evolution is fueled by epigenetic as well as genetic diversity. In chronic lymphocytic leukemia (CLL), intra-tumoral DNA methylation (DNAme) heterogeneity empowers evolution. Here, to comprehensively study the epigenetic dimension of cancer evolution, we integrate DNAme analysis with histone modification mapping and single cell analyses of RNA expression and DNAme in 22 primary CLL and 13 healthy donor B lymphocyte samples. Our data reveal corrupted coherence across different layers of the CLL epigenome. This manifests in decreased mutual information across epigenetic modifications and gene expression attributed to cell-to-cell heterogeneity. Disrupted epigenetic-transcriptional coordination in CLL is also reflected in the dysregulation of the transcriptional output as a function of the combinatorial chromatin states, including incomplete Polycomb-mediated gene silencing. Notably, we observe unexpected co-mapping of typically mutually exclusive activating and repressing histone modifications, suggestive of intra-tumoral epigenetic diversity. Thus, CLL epigenetic diversification leads to decreased coordination across layers of epigenetic information, likely reflecting an admixture of cells with diverging cellular identities. In chronic lymphocytic leukemia (CLL), evolution is driven by transcriptional and epigenetic heterogeneity. Here, the authors integrate epigenomic analyses to show how intra-tumoral epigenetic diversity results in divergent chromatin states in CLL cells, increasing cell-to-cell transcriptional heterogeneity.
Collapse
|
4
|
Development of a Deimmunized Bispecific Immunotoxin dDT2219 against B-Cell Malignancies. Toxins (Basel) 2018; 10:toxins10010032. [PMID: 29316610 PMCID: PMC5793119 DOI: 10.3390/toxins10010032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/16/2017] [Accepted: 12/31/2017] [Indexed: 12/13/2022] Open
Abstract
Diphtheria toxin (DT) related targeted toxins are effective in cancer treatment, but efficacy diminishes in time because of their immunogenic potential and/or former vaccinations. In order to overcome this limitation for DT2219, a promising bispecific targeted toxin which targets CD19 and CD22, we deimmunized the DT moiety, and thereby developed an exciting improved drug (dDT2219) which still has the potential to sufficiently target B-cell malignancies but also limits clearance because of its reduced immunogenicity. The DT moiety was modified by inducing point mutations in prominent positions on the molecular surface. The new engineered dDT2219 was tested for activity, efficacy, and specificity using functional assays, proliferation assays, and flow cytometry. Furthermore, 12 samples of Chronic Lymphatic Leukemia (CLL) patients were used to assess binding. Immunogenicity was determined using a BALB/c mouse model. dDT2219 was efficient and specific against B-cell malignancies such as Bukitt-Lymphoma cell lines Daudi and Raji. dDT2219 showed specific binding on targets and on CLL samples. Intraperitoneal vaccination of immune competent mice showed that even after multiple administrations with increasing doses, induction of neutralizing antibodies was significantly lower in the dDT2219 treated animal group. The new dDT2219 combines potent anti-tumor cell activity with a reduced immunogenicity. With regard to the frequent development of neutralizing antibodies after multiple administrations with immunotoxins, dDT2219 shows promise to overcome this limitation and thus might maintain effectiveness even after multiple treatment cycles.
Collapse
|
5
|
Li Y, Wu S, Bai F. Molecular characterization of circulating tumor cells-from bench to bedside. Semin Cell Dev Biol 2017; 75:88-97. [PMID: 28899718 DOI: 10.1016/j.semcdb.2017.09.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells discovered in cancer patients' peripheral blood that successfully escape from the primary tumor site and/or metastases, struggle to survive in the bloodstream, and have potential for seeding metastases. Numerous methods have been proposed to capture CTCs. The value of CTCs as a means of understanding cancer metastasis and a major form of 'liquid biopsy' has been widely demonstrated. Recently, single-cell molecular analyses of CTCs have provided profound biological insights into tumor heterogeneity, mechanism of metastasis and tumor evolution. In addition, because CTC analysis is non-invasive, CTCs exhibit great potential as biomarkers for assessment of cancer prognosis and therapy response. In this review, we summarize modern technologies for CTC detection and isolation, single-cell genomic/transcriptomic characterization of CTCs, and prospective clinical applications of CTCs. We expect that, after further technical improvements in methods of detection and sequencing, CTC analyses will shed new light on the mechanisms driving cancer metastasis and benefit many cancer patients.
Collapse
Affiliation(s)
- Yanmeng Li
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Science, Peking University, Beijing 100871, China
| | - Shaohan Wu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Science, Peking University, Beijing 100871, China
| | - Fan Bai
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Science, Peking University, Beijing 100871, China.
| |
Collapse
|
6
|
Buxbaum NP, Farthing DE, Maglakelidze N, Lizak M, Merkle H, Carpenter AC, Oliver BU, Kapoor V, Castro E, Swan GA, Dos Santos LM, Bouladoux NJ, Bare CV, Flomerfelt FA, Eckhaus MA, Telford WG, Belkaid Y, Bosselut RJ, Gress RE. In vivo kinetics and nonradioactive imaging of rapidly proliferating cells in graft-versus-host disease. JCI Insight 2017; 2:92851. [PMID: 28614804 DOI: 10.1172/jci.insight.92851] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/16/2017] [Indexed: 12/25/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) offers a cure for cancers that are refractory to chemotherapy and radiation. Most HSCT recipients develop chronic graft-versus-host disease (cGVHD), a systemic alloimmune attack on host organs. Diagnosis is based on clinical signs and symptoms, as biopsies are risky. T cells are central to the biology of cGVHD. We found that a low Treg/CD4+ T effector memory (Tem) ratio in circulation, lymphoid, and target organs identified early and established mouse cGVHD. Using deuterated water labeling to measure multicompartment in vivo kinetics of these subsets, we show robust Tem and Treg proliferation in lymphoid and target organs, while Tregs undergo apoptosis in target organs. Since deuterium enrichment into DNA serves as a proxy for cell proliferation, we developed a whole-body clinically relevant deuterium MRI approach to nonradioactively detect cGVHD and potentially allow imaging of other diseases characterized by rapidly proliferating cells.
Collapse
Affiliation(s)
- Nataliya P Buxbaum
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Donald E Farthing
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | | | - Martin Lizak
- In Vivo NMR Center, National Institute of Neurological Disorders and Stroke
| | - Hellmut Merkle
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke
| | | | - Brittany U Oliver
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Veena Kapoor
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Ehydel Castro
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Gregory A Swan
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Liliane M Dos Santos
- Mucosal Immunology Section, National Institute of Allergy and Infectious Diseases, and
| | - Nicolas J Bouladoux
- Mucosal Immunology Section, National Institute of Allergy and Infectious Diseases, and
| | - Catherine V Bare
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | | | - Michael A Eckhaus
- Diagnostic and Research Services Branch, Office of the Director, NIH, Bethesda, Maryland, USA
| | - William G Telford
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| | - Yasmine Belkaid
- Mucosal Immunology Section, National Institute of Allergy and Infectious Diseases, and
| | - Remy J Bosselut
- Laboratory of Immune Cell Biology, National Cancer Institute
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute
| |
Collapse
|
7
|
Murphy EJ, Neuberg DS, Rassenti LZ, Hayes G, Redd R, Emson C, Li K, Brown JR, Wierda WG, Turner S, Greaves AW, Zent CS, Byrd JC, McConnel C, Barrientos J, Kay N, Hellerstein MK, Chiorazzi N, Kipps TJ, Rai KR. Leukemia-cell proliferation and disease progression in patients with early stage chronic lymphocytic leukemia. Leukemia 2017; 31:1348-1354. [PMID: 28115735 PMCID: PMC5462857 DOI: 10.1038/leu.2017.34] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/23/2016] [Accepted: 01/11/2017] [Indexed: 01/23/2023]
Abstract
The clinical course of patients with recently diagnosed early stage chronic lymphocytic leukemia (CLL) is highly variable. We examined the relationship between CLL-cell birth rate and treatment-free survival (TFS) in 97 patients with recently diagnosed, Rai stage 0-II CLL in a blinded, prospective study, using in vivo 2H2O labeling. Birth rates ranged from 0.07 to 1.31% new cells per day. With median follow-up of 4.0 years, 33 subjects (34%) required treatment by NCI criteria. High-birth rate was observed in 44% of subjects and was significantly associated with shorter TFS, unmutated IGHV status and expression of ZAP70 and of CD38. In multivariable modeling considering age, gender, Rai stage, expression of ZAP70 or CD38, IGHV mutation status and FISH cytogenetics, only CLL-cell birth rate and IGHV mutation status met criteria for inclusion. Hazard ratios were 3.51 (P=0.002) for high-birth rate and 4.93 (P<0.001) for unmutated IGHV. The association between elevated birth rate and shorter TFS was observed in subjects with either mutated or unmutated IGHVs, and the use of both markers was a better predictor of TFS than either parameter alone. Thus, an increased CLL birth rate in early stage disease is a strong predictor of disease progression and earlier treatment.
Collapse
Affiliation(s)
- E J Murphy
- Department of Medicine, University of California, San Francisco, CA, USA
- KineMed Inc., Emeryville, CA, USA
| | - D S Neuberg
- Dana Farber Cancer Institute, Boston, MA, USA
- CLL Research Consortium, San Diego, CA, USA
| | - L Z Rassenti
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Moores Cancer Center, University of California, San Diego, CA, USA
| | - G Hayes
- KineMed Inc., Emeryville, CA, USA
| | - R Redd
- Dana Farber Cancer Institute, Boston, MA, USA
| | - C Emson
- KineMed Inc., Emeryville, CA, USA
| | - K Li
- KineMed Inc., Emeryville, CA, USA
| | - J R Brown
- Dana Farber Cancer Institute, Boston, MA, USA
- CLL Research Consortium, San Diego, CA, USA
| | - W G Wierda
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, M. D. Anderson Cancer Center, Houston, TX, USA
| | - S Turner
- KineMed Inc., Emeryville, CA, USA
| | - A W Greaves
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Moores Cancer Center, University of California, San Diego, CA, USA
| | - C S Zent
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - J C Byrd
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Ohio State University, Columbus, OH, USA
| | | | - J Barrientos
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Hofstra Northwell School of Medicine, Manhasset, NY, USA
| | - N Kay
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - M K Hellerstein
- KineMed Inc., Emeryville, CA, USA
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA, USA
| | - N Chiorazzi
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Hofstra Northwell School of Medicine, Manhasset, NY, USA
- Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - T J Kipps
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Moores Cancer Center, University of California, San Diego, CA, USA
| | - K R Rai
- CLL Research Consortium, San Diego, CA, USA
- Department of Medicine, Hofstra Northwell School of Medicine, Manhasset, NY, USA
- Feinstein Institute for Medical Research, Manhasset, NY, USA
| |
Collapse
|
8
|
Direct in vivo evidence for increased proliferation of CLL cells in lymph nodes compared to bone marrow and peripheral blood. Leukemia 2017; 31:1340-1347. [PMID: 28074063 PMCID: PMC5462849 DOI: 10.1038/leu.2017.11] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/28/2016] [Accepted: 12/13/2016] [Indexed: 12/15/2022]
Abstract
Chronic Lymphocytic Leukemia (CLL) is a progressive malignancy of mature B-cells that involves the peripheral blood (PB), lymph nodes (LNs) and bone marrow (BM). While the majority of CLL cells are in a resting state, small populations of proliferating cells exist; however, the anatomical site of active cell proliferation remains to be definitively determined. Based on findings that CLL cells in LNs have increased expression of B-cell activation genes, we tested the hypothesis that the fraction of “newly born” cells would be highest in the LNs. Using a deuterium oxide (2H) in vivo labeling method in which patients consumed deuterated (heavy) water (2H2O), we determined CLL cell kinetics in concurrently obtained samples from LN, PB, and BM. The LN was identified as the anatomical site harboring the largest fraction of newly born cells, compared to PB and BM. In fact, the calculated birth rate in the LN reached as high a 3.3% of the clone per day. Subdivision of the bulk CLL population by flow cytometry identified the subpopulation with the CXCR4dimCD5bright phenotype as containing the highest proportion of newly born cells within each compartment, including the LN, identifying this subclonal population as an important target for novel treatment approaches.
Collapse
|
9
|
Balakrishnan K, Peluso M, Fu M, Rosin NY, Burger JA, Wierda WG, Keating MJ, Faia K, O'Brien S, Kutok JL, Gandhi V. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia 2015; 29:1811-22. [PMID: 25917267 DOI: 10.1038/leu.2015.105] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/26/2015] [Accepted: 04/07/2015] [Indexed: 11/10/2022]
Abstract
The functional relevance of the B-cell receptor (BCR) and the evolution of protein kinases as therapeutic targets have recently shifted the paradigm for treatment of B-cell malignancies. Inhibition of p110δ with idelalisib has shown clinical activity in chronic lymphocytic leukemia (CLL). The dynamic interplay of isoforms p110δ and p110γ in leukocytes support the hypothesis that dual blockade may provide a therapeutic benefit. IPI-145, an oral inhibitor of p110δ and p110γ isoforms, sensitizes BCR-stimulated and/or stromal co-cultured primary CLL cells to apoptosis (median 20%, n=57; P<0.0001) including samples with poor prognostic markers, unmutated IgVH (n=28) and prior treatment (n=15; P<0.0001). IPI-145 potently inhibits the CD40L/IL-2/IL-10 induced proliferation of CLL cells with an IC50 in sub-nanomolar range. A corresponding dose-responsive inhibition of pAKT(Ser473) is observed with an IC50 of 0.36 nM. IPI-145 diminishes the BCR-induced chemokines CCL3 and CCL4 secretion to 17% and 37%, respectively. Pre-treatment with 1 μM IPI-145 inhibits the chemotaxis toward CXCL12; reduces pseudoemperipolesis to median 50%, inferring its ability to interfere with homing capabilities of CLL cells. BCR-activated signaling proteins AKT(Ser473), BAD(Ser112), ERK(Thr202/Tyr204) and S6(Ser235/236) are mitigated by IPI-145. Importantly, for clinical development in hematological malignancies, IPI-145 is selective to CLL B cells, sparing normal B- and T-lymphocytes.
Collapse
Affiliation(s)
- K Balakrishnan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Peluso
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - M Fu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N Y Rosin
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Faia
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - S O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J L Kutok
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - V Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Weinkove R, Brooks CR, Carter JM, Hermans IF, Ronchese F. Efficient depletion of chronic lymphocytic leukemia B cells using serial rounds of immunomagnetic depletion. J Immunol Methods 2013; 396:152-6. [PMID: 23911312 DOI: 10.1016/j.jim.2013.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/16/2013] [Accepted: 07/22/2013] [Indexed: 11/19/2022]
Abstract
Functional studies of cellular immunity in patients with leukemia often require separation of leukemic cells from other peripheral blood mononuclear cells (PBMCs). This can pose a challenge when the number of leukemic cells is very high, such as in untreated patients with chronic lymphocytic leukemia (CLL). We found that when leukemia cell frequency was very high, anti-CD19 coated immunomagnetic beads did not thoroughly deplete B cells when used according to manufacturer's instructions. In this study, we depleted leukemic B cells using a modified protocol comprising serial rounds of depletion using immunomagnetic beads at reduced bead to cell ratios. This resulted in more effective B cell depletion with the use of fewer immunomagnetic beads, and without affecting viability or yield of non-B cells. CD19- PBMC subsets were retained, and serial depletion rounds did not activate T cells and monocytes. The positively isolated CLL cells were of high purity and were available for downstream analysis. This is a convenient and cost-effective method to enable in vitro analysis of immunocompetent cells from patients with leukemia.
Collapse
Affiliation(s)
- Robert Weinkove
- Malaghan Institute of Medical Research, Wellington, New Zealand; Wellington Blood & Cancer Centre, Capital & Coast District Health Board, Wellington, New Zealand; Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, New Zealand.
| | | | | | | | | |
Collapse
|
11
|
Arya SK, Lim B, Rahman ARA. Enrichment, detection and clinical significance of circulating tumor cells. LAB ON A CHIP 2013; 13:1995-2027. [PMID: 23625167 DOI: 10.1039/c3lc00009e] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Circulating Tumor Cells (CTCs) are shed from primary or secondary tumors into blood circulation. Accessing and analyzing these cells provides a non-invasive alternative to tissue biopsy. CTCs are estimated to be as few as 1 cell among a few million WBCs and few billion RBCs in 1 ml of patient blood and are rarely found in healthy individuals. CTCs are FDA approved for prognosis of the major cancers, namely, Breast, Colon and Prostate. Currently, more than 400 clinical trials are ongoing to establish their clinical significance beyond prognosis, such as, therapy selection and companion diagnostics. Understanding the clinical relevance of CTCs typically involves isolation, detection and molecular characterization of cells, ideally at single cell level. The need for highly reliable, standardized and robust methodologies for isolating and analyzing CTCs has been widely expressed by clinical thought leaders. In the last decade, numerous academic and commercial technology platforms for isolation and analysis of CTCs have been reported. A recent market report highlighted the presence of more than 100 companies offering products and services related to CTCs. This review aims to capture the state of the art and examines the technical merits and limitations of contemporary technologies for clinical use.
Collapse
Affiliation(s)
- Sunil K Arya
- Bioelectronics Programme, Institute of Microelectronics, A*STAR (Agency for Science, Technology and Research), 11 Science Park Road, Singapore Science Park II, Singapore 117685.
| | | | | |
Collapse
|
12
|
Network properties derived from deep sequencing of human B-cell receptor repertoires delineate B-cell populations. Genome Res 2013; 23:1874-84. [PMID: 23742949 PMCID: PMC3814887 DOI: 10.1101/gr.154815.113] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The adaptive immune response selectively expands B- and T-cell clones following antigen recognition by B- and T-cell receptors (BCR and TCR), respectively. Next-generation sequencing is a powerful tool for dissecting the BCR and TCR populations at high resolution, but robust computational analyses are required to interpret such sequencing. Here, we develop a novel computational approach for BCR repertoire analysis using established next-generation sequencing methods coupled with network construction and population analysis. BCR sequences organize into networks based on sequence diversity, with differences in network connectivity clearly distinguishing between diverse repertoires of healthy individuals and clonally expanded repertoires from individuals with chronic lymphocytic leukemia (CLL) and other clonal blood disorders. Network population measures defined by the Gini Index and cluster sizes quantify the BCR clonality status and are robust to sampling and sequencing depths. BCR network analysis therefore allows the direct and quantifiable comparison of BCR repertoires between samples and intra-individual population changes between temporal or spatially separated samples and over the course of therapy.
Collapse
|
13
|
Abstract
The detection of circulating tumor cells (CTC) aids in diagnosis of disease, prognosis, disease recurrence, and therapeutic response. The molecular aspects of metastasis are reviewed including its relevance in the identification and characterization of putative markers that may be useful in the detection thereof. Also discussed are methods for CTC enrichment using molecular strategies. The clinical application of CTC in the metastatic disease process is also summarized.
Collapse
|
14
|
Song Y, Wang Z, Yang L. [Advances in research on circulating tumor cells in lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2012; 15:612-8. [PMID: 23075687 PMCID: PMC5999834 DOI: 10.3779/j.issn.1009-3419.2012.10.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
转移和复发是肺癌患者死亡的主要原因。研究发现循环肿瘤细胞(circulating tumor cells, CTCs)在肺癌转移和复发中起着重要作用。而且随着靶向治疗的不断进步,对于晚期无法取得肺癌实体组织的患者,CTCs作为一种肺癌组织替代物可以决定治疗方案。所以CTCs在早期发现肺癌患者的微转移、检测肿瘤复发、评估预后和选择个体化治疗方案方面有着重要作用。本文针对CTCs的研究进展及肺癌领域的应用进行综述。
Collapse
Affiliation(s)
- Yingjian Song
- Department of Thoracic Surgery, Shenzhen People's Hospital, the Second Clinical Medicine College of Jinan University,
Shenzhen 518020, China
| | | | | |
Collapse
|
15
|
Kowdley G, Srikantan S, Abdelmohsen K, Gorospe M, Khan J. Molecular biology techniques for the surgeon. World J Surg Proced 2012; 2:5-15. [DOI: 10.5412/wjsp.v2.i2.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
New technologies are constantly being introduced into the medical and surgical fields. These technologies come in the form of newer medicines, imaging methods and prognostic tools, among others, and allow clinicians to make more rational and informed decisions on the care of their patients. Many of these technologies utilize advanced techniques which are at the forefront of many research fields and represent a transition of bench advances into the clinical realm. This review will highlight four technologies that are at the forefront in the treatment of oncology patients treated by surgeons on a daily basis. Circulating tumor cells, microarray analysis, proteomic studies and rapid sequencing technologies will be highlighted. These technologies will be reviewed and their potential use in the care of surgical patients will be discussed.
Collapse
|
16
|
Sun YF, Yang XR, Zhou J, Qiu SJ, Fan J, Xu Y. Circulating tumor cells: advances in detection methods, biological issues, and clinical relevance. J Cancer Res Clin Oncol 2011; 137:1151-73. [PMID: 21681690 DOI: 10.1007/s00432-011-0988-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 05/26/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND Circulating tumor cells (CTCs) have long been considered a reflection of tumor aggressiveness. Hematogenous spreading of CTCs from a primary tumor is a crucial step in the metastasis cascade, which leads ultimately to the formation of overt metastases. However, owing to the rarity of CTCs in peripheral blood, detecting these cells requires methods combined with high sensitivity and specificity, which sets tremendous challenges for the implementation of these assays into clinical routine. METHODS Generally, CTCs detection methods are composed of the following two steps: enrichment (isolation) process (morphological and immunological techniques) and detection (identification) process (cytometric and nucleic acid techniques), which may or may not be separate from enrichment. Genetic and molecular characterization of CTCs carried out by fluorescent in situ hybridization (FISH), comparative genomic hybridization (CGH), PCR-based techniques, and biomarker immunofluorescent staining extract more information about malignant profile, metastatic potential of CTCs, and the extent to which CTCs are genetically identical to the primary tumor. RESULTS Recent technical advances made it possible to detect CTCs. The efficacy of circulating tumor cell (CTC) detection among patients with solid malignancy has been investigated, which shows great potential to become a tool for real-time parameter of prognosis and serve as an early marker to assess the therapeutic response in overt cancers. Improvements in detection and characterization of CTCs will hopefully lead to refinement of clinical management of cancer patients. CONCLUSION This review addresses the majority of assays that have been published thus far, including the enrichment and detection steps and the markers used in these assays, accompanied by some biological issues of CTC and the results of clinical application harvested.
Collapse
Affiliation(s)
- Yun-Fan Sun
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhong Shan Hospital, Fudan University, 136 Yi Xue Yuan Road, Shanghai 200032, People's Republic of China
| | | | | | | | | | | |
Collapse
|
17
|
Friel AM, Corcoran C, Crown J, O'Driscoll L. Relevance of circulating tumor cells, extracellular nucleic acids, and exosomes in breast cancer. Breast Cancer Res Treat 2010; 123:613-25. [PMID: 20549336 DOI: 10.1007/s10549-010-0980-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 06/02/2010] [Indexed: 12/17/2022]
Abstract
Early detection of cancer is vital to improved overall survival rates. At present, evidence is accumulating for the clinical value of detecting occult tumor cells in peripheral blood, plasma, and serum specimens from cancer patients. Both molecular and cellular approaches, which differ in sensitivity and specificity, have been used for such means. Circulating tumor cells and extracellular nucleic acids have been detected within blood, plasma, and sera of cancer patients. As the presence of malignant tumors are clinically determined and/or confirmed upon biopsy procurement-which in itself may have detrimental effects in terms of stimulating cancer progression/metastases-minimally invasive methods would be highly advantageous to the diagnosis and prognosis of breast cancer and the subsequent tailoring of targeted treatments for individuals, if reliable panels of biomarkers suitable for such an approach exist. Herein, we review the current advances made in the detection of such circulating tumor cells and nucleic acids, with particular emphasis on extracellular nucleic acids, specifically extracellular mRNAs and discuss their clinical relevance.
Collapse
Affiliation(s)
- Anne M Friel
- School of Pharmacy and Pharmaceutical Sciences & Molecular Therapeutics for Cancer Ireland, Trinity College Dublin, Dublin 2, Ireland
| | | | | | | |
Collapse
|