Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Drug Deliv ; 28(1): 800-813, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33866918

ABSTRACT

Cancer immunotherapy is a strategy that is moving to the frontier of cancer treatment in the current decade. In this study, we show evidence that 3-(2-nitrophenyl) propionic acid-paclitaxel nanoparticles (NPPA-PTX NPs), act as immunogenic cell death (ICD) inducers, stimulating an antitumor response which results in synergistic antitumor activity by combining anti-PD-L1 antibody (aPD-L1) in vivo. To investigate the antitumor immunity induced by NPPA-PTX NPs, the expression of both ICD marker calreticulin (CRT) and high mobility group box 1 (HMGB1) were analyzed. In addition, the antitumor activity of NPPA-PTX NPs combined with aPD-L1 in vivo was also investigated. The immune response was also measured through quantitation of the infiltration of T cells and the secretion of pro-inflammatory cytokines. The results demonstrate that NPPA-PTX NPs induce ICD of MDA-MB-231 and 4T1 cells through upregulation of CRT and HMGB1, reactivating the antitumor immunity via recruitment of infiltrating CD3+, CD4+, CD8+ T cells, secreting IFN-γ, TNF-α, and the enhanced antitumor activity by combining with aPD-L1. These data suggest that the combined therapy has a synergistic antitumor activity and has the potential to be developed into a novel therapeutic regimen for cancer patients.


Subject(s)
Albumins/pharmacology , Antineoplastic Agents/pharmacology , Immunogenic Cell Death/drug effects , Inflammation Mediators/metabolism , Nanoparticles/chemistry , Paclitaxel/pharmacology , Albumins/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , B7-H1 Antigen/immunology , Calreticulin/drug effects , Cell Line, Tumor , Female , HMGB1 Protein/drug effects , Humans , Mice , Mice, Inbred BALB C , Paclitaxel/administration & dosage , Propionates/chemistry , Up-Regulation , Xenograft Model Antitumor Assays
2.
Stroke ; 49(12): 3020-3029, 2018 12.
Article in English | MEDLINE | ID: mdl-30571407

ABSTRACT

Background and Purpose- Heme and iron are considered to be key factors responsible for secondary insults after intracerebral hemorrhage (ICH). Our previous study showed that LRP1 (low-density lipoprotein receptor-related protein-1)-Hx (hemopexin) facilitates removal of heme. The TLR7 (Toll-like receptor 7)-BTK (Bruton tyrosine kinase)-CRT (calreticulin) pathway regulates the expression of LRP1-Hx. This study is designed to clarify whether TLR7 activation facilitates heme scavenging and to establish the potential role of the BTK-CRT-LRP1-Hx signaling pathway in the pathophysiology of ICH. Methods- ICH was induced by stereotactic, intrastriatal injection of type VII collagenase. Mice received TLR7 agonist (imiquimod) via intraperitoneal injection after ICH induction. TLR7 inhibitor (ODN2088), BTK inhibitor (LFM-A13), and CRT agonist (thapsigargin) were given in different groups to further evaluate the underlying pathway. Mice were randomly divided into sham, ICH+vehicle (normal saline), ICH+Imiquimod (2.5, 5, and 10 µg/g), ICH+ODN2088, ICH+LFM-A13, ICH+thapsigargin, and ICH+ODN2088+thapsigargin. Imiquimod was administered twice daily starting at 6 hours after ICH; ODN2088 was administered by intracerebroventricular injection at 30 minutes, and LFM-A13 or thapsigargin was administered by intraperitoneal injection at 3 hours after ICH induction. Neurological scores, cognitive abilities, as well as brain edema, blood-brain barrier permeability, hemoglobin level, brain expression of TLR7/BTK/CRT/LRP1/Hx were analyzed. Results- Low dosage imiquimod significantly attenuated hematoma volume, brain edema, BBB permeability, and neurological deficits after ICH. Imiquimod also increased protein expressions of TLR7, BTK, CRT, LRP1, and Hx; ODN2088 reduced TLR7, BTK, CRT, LRP1, and Hx expressions. Conclusions- TLR7 plays an important role in heme scavenging after ICH by modulating the BTK-CRT-LRP1-Hx pathway. TLR7 may offer protective effects by promoting heme resolution and reduction of brain edema after ICH.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Brain/metabolism , Calreticulin/metabolism , Cerebral Hemorrhage/metabolism , Heme/metabolism , Hemopexin/metabolism , Membrane Glycoproteins/metabolism , Receptors, LDL/metabolism , Toll-Like Receptor 7/metabolism , Tumor Suppressor Proteins/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/drug effects , Amides/pharmacology , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain Edema/metabolism , Calreticulin/agonists , Calreticulin/drug effects , Enzyme Inhibitors/pharmacology , Hemopexin/drug effects , Imiquimod/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Membrane Glycoproteins/agonists , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/drug effects , Mice , Nitriles/pharmacology , Oligodeoxyribonucleotides/pharmacology , Receptors, LDL/drug effects , Signal Transduction , Thapsigargin/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/drug effects , Tumor Suppressor Proteins/drug effects
3.
Nat Med ; 22(6): 624-31, 2016 06.
Article in English | MEDLINE | ID: mdl-27135741

ABSTRACT

Cetuximab is a monoclonal antibody that is effective in the treatment of metastatic colorectal cancer (mCRC). Cetuximab blocks epidermal growth factor receptor (EGFR)-ligand interaction and inhibits downstream RAS-ERK activation. However, only some activating mutations in RAS affect cetuximab efficacy, and it is not clear what else mediates treatment success. Here we hypothesized that cetuximab induces immunogenic cell death (ICD) that activates a potent antitumor response. We found that cetuximab, in combination with chemotherapy, fostered ICD in CRC cells, which we measured via the endoplasmic reticulum (ER) stress response and an increase in phagocytosis by dendritic cells. ICD induction depended on the mutational status of the EGFR signaling pathway and on the inhibition of the splicing of X-box binding protein 1 (XBP1), an unfolded protein response (UPR) mediator. We confirmed the enhanced immunogenicity elicited by cetuximab in a mouse model of human EGFR-expressing CRC. Overall, we demonstrate a new, immune-related mechanism of action of cetuximab that may help to tailor personalized medicine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/drug effects , Cetuximab/pharmacology , Colorectal Neoplasms/immunology , Endoplasmic Reticulum Stress/drug effects , Phagocytosis/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Calreticulin/drug effects , Calreticulin/metabolism , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Death/immunology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Endoplasmic Reticulum Stress/immunology , Fluorouracil/administration & dosage , HCT116 Cells , HT29 Cells , Humans , Indoles/pharmacology , Irinotecan , Leucovorin/administration & dosage , Mice , Panitumumab , Phagocytosis/immunology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Unfolded Protein Response , Vemurafenib , X-Box Binding Protein 1/drug effects , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism
5.
Arthritis Rheumatol ; 67(8): 2061-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25892196

ABSTRACT

OBJECTIVE: The mechanisms underlying bone damage in rheumatoid arthritis (RA) are incompletely understood. We recently identified the shared epitope (SE), an HLA-DRB1-coded 5-amino acid sequence motif carried by the majority of RA patients as a signal transduction ligand that interacts with cell surface calreticulin and accelerates osteoclast (OC)-mediated bone damage in collagen-induced arthritis (CIA). Given the role of the SE/calreticulin pathway in arthritis-associated bone damage, we sought to determine the therapeutic targetability of calreticulin. METHODS: A library of backbone-cyclized peptidomimetic compounds, all carrying an identical core DKCLA sequence, was synthesized. The ability of these compounds to inhibit SE-activated signaling and OC differentiation was tested in vitro. The effect on disease severity and OC-mediated bone damage was studied by weekly intraperitoneal administration of the compounds to DBA/1 mice with CIA. RESULTS: Two members of the peptidomimetics library were found to have SE-antagonistic effects and antiosteoclast differentiation effects at picomolar concentrations in vitro. The lead mimetic compound, designated HS(4-4)c Trp, potently ameliorated arthritis and bone damage in vivo when administered in picogram doses to mice with CIA. Another mimetic analog, designated HS(3-4)c Trp, was found to lack activity, both in vitro and in vivo. The differential activity of the 2 analogs depended on minor differences in their respective ring sizes and correlated with distinctive geometry when computationally docked to the SE binding site on calreticulin. CONCLUSION: These findings identify calreticulin as a novel therapeutic target in erosive arthritis and provide sound rationale and early structure/activity relationships for future drug design.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone and Bones/drug effects , Calreticulin/drug effects , Histocompatibility Antigens Class II/drug effects , Osteoclasts/drug effects , Peptides/pharmacology , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Bone and Bones/metabolism , Calreticulin/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Epitopes , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , In Vitro Techniques , Ligands , Mice , Molecular Docking Simulation , Osteoclasts/metabolism , Peptide Library , Signal Transduction/drug effects
6.
Cancer Chemother Pharmacol ; 66(3): 611-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20221600

ABSTRACT

PURPOSE: It has recently been recognised that anticancer chemotherapy can elicit an immunogenic form of apoptosis characterised by the exposure of calreticulin (CRT) on the surface of dying tumour cells, entailing an immune response that contributes to the therapeutic outcome. CRT exposure has been found to be induced by anthracyclins and oxaliplatin, but not by other proapoptotic antineoplastic agents including etoposide, camptothecin and cisplatin. In this study, we examined the histone deacetylase inhibitor vorinostat for its capability to stimulate CRT exposure in tumour cells. METHODS: Childhood tumour cells, i.e. the brain tumour cell lines PFSK and DAOY and the Ewing's sarcoma cell line CADO-ES-1, were treated with vorinostat, and CRT exposure was determined by flow cytometric analysis of CRT immunofluorescence. Combination effects of vorinostat/TRAIL and vorinostat/bortezomib were also assessed. RESULTS: Vorinostat treatment induced CRT exposure in PFSK and DAOY cells, but not in caspase-8-deficient CADO-ES-1 cells. CRT exposure could be prevented by the pan-caspase inhibitor z-VAD-fmk and by brefeldin A, an inhibitor of Golgi-mediated transport. CONCLUSION: Vorinostat has the capacity to elicit CRT exposure, suggesting its usefulness as immunogenic antitumour agent.


Subject(s)
Brain Neoplasms/drug therapy , Calreticulin/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Brain Neoplasms/pathology , Calreticulin/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Child , Flow Cytometry , Fluorescent Antibody Technique/methods , Humans , Pyrazines/pharmacology , Vorinostat
7.
Cell Death Dis ; 1: e104, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21368877

ABSTRACT

Experiments performed in mice revealed that anthracyclines stimulate immunogenic cell death that is characterized by the pre-apoptotic exposure of calreticulin (CRT) on the surface of dying tumor cells. Here, we determined whether CRT exposure at the cell surface (ecto-CRT) occurs in human cancer in response to anthracyclines in vivo, focusing on acute myeloid leukemia (AML), which is currently treated with a combination of aracytine and anthracyclines. Most of the patients benefit from the induction chemotherapy but relapse within 1-12 months. In this study, we investigated ecto-CRT expression on malignant blasts before and after induction chemotherapy. We observed that leukemic cells from some patients exhibited ecto-CRT regardless of chemotherapy and that this parameter was not modulated by in vivo chemotherapy. Ecto-CRT correlated with the presence of phosphorylated eIF2α within the blasts, in line with the possibility that CRT exposure results from an endoplasmic reticulum stress response. Importantly, high levels of ecto-CRT on malignant myeloblasts positively correlated with the ability of autologous T cells to secrete interferon-γ on stimulation with blast-derived dendritic cell. We conclude that the presence of ecto-CRT on leukemia cells facilitates cellular anticancer immune responses in AML patients.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Calreticulin/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Calreticulin/drug effects , Cohort Studies , Dendritic Cells/physiology , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Immunity, Cellular/drug effects , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Phagocytosis/drug effects , Phosphorylation , Survival Analysis , T-Lymphocytes/immunology
8.
J Neurochem ; 106(1): 333-46, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18384645

ABSTRACT

In Parkinson's disease, oxidative stress is implicated in protein misfolding and aggregation, which may activate the unfolded protein response by the endoplasmic reticulum (ER). Dopamine (DA) can initiate oxidative stress via H(2)O(2) formation by DA metabolism and by oxidation into DA quinone. We have previously shown that DA quinone induces oxidative protein modification, mitochondrial dysfunction in vitro, and dopaminergic cell toxicity in vivo and in vitro. In this study, we used cysteine- and lysine-reactive fluorescent dyes with 2D difference in-gel electrophoresis, mass spectrometry, and peptide mass fingerprint analysis to identify proteins in PC12 cell mitochondrial-enriched fractions that were altered in abundance following DA exposure (150 muM, 16 h). Quantitative changes in proteins labeled with fluorescent dyes indicated increases in a subset of proteins after DA exposure: calreticulin, ERp29, ERp99, Grp58, Grp78, Grp94 and Orp150 (149-260%), and decreased levels of aldolase A (39-42%). Changes in levels of several proteins detected by 2D difference in-gel electrophoresis were confirmed by western blot. Using this unbiased proteomics approach, our findings demonstrated that in PC12 cells, DA exposure leads to a cellular response indicative of ER stress prior to the onset of cell death, providing a potential link between DA and the unfolded protein response in the pathogenesis of Parkinson's disease.


Subject(s)
Dopamine/metabolism , Endoplasmic Reticulum/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Heat-Shock Proteins/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Animals , Calreticulin/drug effects , Calreticulin/metabolism , Dopamine/pharmacology , Down-Regulation/drug effects , Down-Regulation/physiology , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum/drug effects , Fluorescent Dyes , Fructose-Bisphosphate Aldolase/drug effects , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/drug effects , Mass Spectrometry , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Molecular Chaperones/drug effects , Molecular Chaperones/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , PC12 Cells , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Protein Disulfide-Isomerases/drug effects , Protein Disulfide-Isomerases/metabolism , Protein Folding , Proteins/drug effects , Proteins/metabolism , Rats , Up-Regulation/drug effects , Up-Regulation/physiology
9.
Cell Res ; 15(7): 523-31, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16045815

ABSTRACT

The non-classical HLA class I antigen HLA-G is an immune modulator which inhibits the functions of T cells, NK cells, and the Dendritic cells (DC). As a result, HLA-G expression in malignant cells may provide them with a mechanism to escape the immune surveillance. In melanoma, HLA-G antigen expression has been found in 30% of surgically removed lesions but in less than 1% of established cell lines. One possible mechanism underlying the differential HLA-G expression in vivo and in vitro is that the HLA-G gene is epigenetically repressed in melanoma cells in vitro. To test this hypothesis, we treated the HLA-G negative melanoma cell line OCM-1A with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-AC) and analyzed whether HLA-G expression can be restored. Our data strongly suggest that HLA-G is silenced as a result of CpG hypermethylation within a 5' regulatory region encompassing 220 bp upstream of the start codon. After treatment, HLA-G mRNA expression was dramatically increased. Western blot and flow cytometry showed that HLA-G protein was induced. Interestingly, HLA-G cell surface expression on the 5-AC treated OCM-1A cells is much less than that on the HLA-G positive JEG-3 cells while a similar amount of total HLA-G was observed. Possible mechanisms for the difference were analyzed in the study such as cell cold-treatment, peptide loading and antigen processing machinery components (APM) as well as beta2 microglobulin (beta2-m) expression. Data revealed that the APM component calreticulin might be involved in the lower HLA-G surface expression on OCM-1A cells. Taken together, our results indicated that DNA methylation is an important epigenetic mechanism by which HLA-G antigen expression is modulated in melanoma cells in vitro. Furthermore, to the first time, we hypothesized that the deficiency of calreticulin might be involved in the low HLA-G surface expression on the 5-AC treated OCM-1A cells.


Subject(s)
Azacitidine/analogs & derivatives , Choriocarcinoma/metabolism , Gene Expression Regulation, Neoplastic , HLA Antigens/genetics , HLA Antigens/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Melanoma/metabolism , Azacitidine/pharmacology , Calreticulin/drug effects , Calreticulin/metabolism , Cell Line, Tumor , Decitabine , Gene Expression Regulation, Neoplastic/drug effects , HLA Antigens/drug effects , HLA-G Antigens , Histocompatibility Antigens Class I/drug effects , Humans , Methylation , Promoter Regions, Genetic , Transcription, Genetic
10.
Biochem J ; 371(Pt 1): 61-9, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12519072

ABSTRACT

Overexpression of the conserved Ca(2+)-binding proteins calreticulin and calsequestrin impairs cardiac function, leading to premature death. Calreticulin is vital for embryonic development, but also impairs glucocorticoid action. Glucocorticoid overexposure during late fetal life causes intra-uterine growth retardation and programmed hypertension in adulthood. To determine whether intra-uterine growth retardation or programmed hypertension was associated with altered calreticulin or calsequestrin expression, effects of prenatal glucocorticoid overexposure (maternal dexamethasone treatment on days 15-21 of pregnancy) were examined during fetal life and postnatal development until adulthood (24 weeks). Dexamethasone (100 or 200 microg/kg of maternal body weight) was administered via osmotic pump. Calreticulin was detected as a 55 kDa band and calsequestrin as 55 and 63 kDa bands in 21 day fetal hearts. Only the 55 kDa calsequestrin band was detected postnatally. Prenatal glucocorticoid overexposure at the higher dose decreased calreticulin protein expression (26%; P <0.05) but increased calsequestrin protein expression, both 55 and 63 kDa bands, by 87% ( P <0.01) and 78% ( P <0.01); only the 55 kDa calsequestrin band was increased at the lower dose (66%; P <0.05). Offspring of dams treated at the lower dexamethasone dose were studied further. In control offspring, cardiac calreticulin protein expression declined between 2 and 3 weeks of age, and remained suppressed until adulthood. Cardiac calsequestrin protein expression increased 2-fold between fetal day 21 and postnatal day 1 and continued to increase until adulthood, at which time it was 3.4-fold higher ( P <0.001). Prenatal dexamethasone exposure minimally affected postnatal calsequestrin protein expression, but the postnatal decline in calreticulin protein expression was abrogated and calreticulin protein expression in adulthood was 2.2-fold increased ( P <0.001) compared with adult controls. In view of the known associations between cardiac calreticulin overexpression and impaired cardiac function, targeted up-regulation of calreticulin may contribute to the increased risk of adult heart disease introduced as a result of prenatal overexposure to glucocorticoids.


Subject(s)
Calreticulin/metabolism , Calsequestrin/metabolism , Glucocorticoids/adverse effects , Heart/drug effects , Heart/growth & development , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn , Calreticulin/drug effects , Calsequestrin/drug effects , Dexamethasone/adverse effects , Embryonic and Fetal Development/drug effects , Female , Fetal Growth Retardation/chemically induced , Gestational Age , Heart/embryology , Myocardium/metabolism , Pregnancy , Rats , Rats, Wistar
11.
Cancer Res ; 63(1): 100-6, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12517784

ABSTRACT

Thiopurine treatment of human leukemia cells deficient in components of the mismatch repair system (Nalm6) initiated apoptosis after incorporation into DNA, as revealed by caspase activation and terminal deoxynucleotidyl transferase-mediated nick end labeling assay. To elucidate the cellular sensor(s) responsible for recognition of DNA damage in cells with an inactive mismatch repair system, we isolated a multiprotein nuclear complex that preferentially binds DNA with thioguanine incorporated. The components of this nuclear multiprotein complex, as identified by protein mass spectroscopy, included high mobility group proteins 1 and 2 (HMGB1, HMGB2), heat shock protein HSC70, protein disulfide isomerase ERp60, and glyceraldehyde 3-phosphate dehydrogenase. The same complex was also shown to bind synthetic oligodeoxyribonucleotide duplexes containing the nonnatural nucleosides 1-beta-D-arabinofuranosylcytosine or 5-fluoro-2'-deoxyuridine. Fibroblast cell line derived from Hmgb1(-/-) murine embryos had decreased sensitivity to thiopurines, with an IC(50) 10-fold greater than Hmgb1-proficient cells (P < 0.0001) and exhibited comparable sensitivity to vincristine, a cytotoxic drug that is not incorporated into DNA. These findings indicate that the HMGB1-HMGB2-HSC70-ERp60-glyceraldehyde 3-phosphate dehydrogenase complex detects changes in DNA structure caused by incorporation of nonnatural nucleosides and is a determinant of cell sensitivity to such DNA modifying chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Calreticulin/metabolism , Cell Survival/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HSP70 Heat-Shock Proteins/metabolism , High Mobility Group Proteins/metabolism , Nuclear Proteins/metabolism , Base Sequence , Calreticulin/drug effects , Cell Cycle/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , HSP70 Heat-Shock Proteins/drug effects , High Mobility Group Proteins/drug effects , Humans , Kinetics , Leukemia, B-Cell , Macromolecular Substances , Multiprotein Complexes , Nuclear Proteins/drug effects , S Phase/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...