Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cardiovasc Res ; 117(9): 2125-2136, 2021 07 27.
Article in English | MEDLINE | ID: mdl-33002105

ABSTRACT

AIMS: Stem cell therapy has shown promise for treating myocardial infarction via re-muscularization and paracrine signalling in both small and large animals. Non-human primates (NHPs), such as rhesus macaques (Macaca mulatta), are primarily utilized in preclinical trials due to their similarity to humans, both genetically and physiologically. Currently, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are delivered into the infarcted myocardium by either direct cell injection or an engineered tissue patch. Although both approaches have advantages in terms of sample preparation, cell-host interaction, and engraftment, how the iPSC-CMs respond to ischaemic conditions in the infarcted heart under these two different delivery approaches remains unclear. Here, we aim to gain a better understanding of the effects of hypoxia on iPSC-CMs at the transcriptome level. METHODS AND RESULTS: NHP iPSC-CMs in both monolayer culture (2D) and engineered heart tissue (EHT) (3D) format were exposed to hypoxic conditions to serve as surrogates of direct cell injection and tissue implantation in vivo, respectively. Outcomes were compared at the transcriptome level. We found the 3D EHT model was more sensitive to ischaemic conditions and similar to the native in vivo myocardium in terms of cell-extracellular matrix/cell-cell interactions, energy metabolism, and paracrine signalling. CONCLUSION: By exposing NHP iPSC-CMs to different culture conditions, transcriptome profiling improves our understanding of the mechanism of ischaemic injury.


Subject(s)
Cell Differentiation , Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Myocardial Ischemia/genetics , Myocytes, Cardiac/metabolism , Tissue Engineering , Transcriptome , Animals , Cell Hypoxia , Cell-Matrix Junctions , Cells, Cultured , Energy Metabolism , Gene Regulatory Networks , Heart Rate , Induced Pluripotent Stem Cells/pathology , Macaca mulatta , Male , Mice, SCID , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Paracrine Communication , Phenotype
2.
Stem Cell Reports ; 10(2): 422-435, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29398480

ABSTRACT

Non-human primates (NHPs) can serve as a human-like model to study cell therapy using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). However, whether the efficacy of NHP and human iPSC-CMs is mechanistically similar remains unknown. To examine this, RNU rats received intramyocardial injection of 1 × 107 NHP or human iPSC-CMs or the same number of respective fibroblasts or PBS control (n = 9-14/group) at 4 days after 60-min coronary artery occlusion-reperfusion. Cardiac function and left ventricular remodeling were similarly improved in both iPSC-CM-treated groups. To mimic the ischemic environment in the infarcted heart, both cultured NHP and human iPSC-CMs underwent 24-hr hypoxia in vitro. Both cells and media were collected, and similarities in transcriptomic as well as metabolomic profiles were noted between both groups. In conclusion, both NHP and human iPSC-CMs confer similar cardioprotection in a rodent myocardial infarction model through relatively similar mechanisms via promotion of cell survival, angiogenesis, and inhibition of hypertrophy and fibrosis.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Stem Cell Transplantation , Animals , Cell Differentiation , Cell Hypoxia/physiology , Cell Survival/physiology , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/cytology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Primates , Rats
3.
Sci Transl Med ; 9(377)2017 02 15.
Article in English | MEDLINE | ID: mdl-28202772

ABSTRACT

Tyrosine kinase inhibitors (TKIs), despite their efficacy as anticancer therapeutics, are associated with cardiovascular side effects ranging from induced arrhythmias to heart failure. We used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), generated from 11 healthy individuals and 2 patients receiving cancer treatment, to screen U.S. Food and Drug Administration-approved TKIs for cardiotoxicities by measuring alterations in cardiomyocyte viability, contractility, electrophysiology, calcium handling, and signaling. With these data, we generated a "cardiac safety index" to reflect the cardiotoxicities of existing TKIs. TKIs with low cardiac safety indices exhibit cardiotoxicity in patients. We also derived endothelial cells (hiPSC-ECs) and cardiac fibroblasts (hiPSC-CFs) to examine cell type-specific cardiotoxicities. Using high-throughput screening, we determined that vascular endothelial growth factor receptor 2 (VEGFR2)/platelet-derived growth factor receptor (PDGFR)-inhibiting TKIs caused cardiotoxicity in hiPSC-CMs, hiPSC-ECs, and hiPSC-CFs. With phosphoprotein analysis, we determined that VEGFR2/PDGFR-inhibiting TKIs led to a compensatory increase in cardioprotective insulin and insulin-like growth factor (IGF) signaling in hiPSC-CMs. Up-regulating cardioprotective signaling with exogenous insulin or IGF1 improved hiPSC-CM viability during cotreatment with cardiotoxic VEGFR2/PDGFR-inhibiting TKIs. Thus, hiPSC-CMs can be used to screen for cardiovascular toxicities associated with anticancer TKIs, and the results correlate with clinical phenotypes. This approach provides unexpected insights, as illustrated by our finding that toxicity can be alleviated via cardioprotective insulin/IGF signaling.


Subject(s)
Cardiotoxicity/pathology , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/metabolism , Protein Kinase Inhibitors/toxicity , Biomarkers/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Sarcomeres/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Nat Med ; 22(5): 547-56, 2016 05.
Article in English | MEDLINE | ID: mdl-27089514

ABSTRACT

Doxorubicin is an anthracycline chemotherapy agent effective in treating a wide range of malignancies, but it causes a dose-related cardiotoxicity that can lead to heart failure in a subset of patients. At present, it is not possible to predict which patients will be affected by doxorubicin-induced cardiotoxicity (DIC). Here we demonstrate that patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can recapitulate the predilection to DIC of individual patients at the cellular level. hiPSC-CMs derived from individuals with breast cancer who experienced DIC were consistently more sensitive to doxorubicin toxicity than hiPSC-CMs from patients who did not experience DIC, with decreased cell viability, impaired mitochondrial and metabolic function, impaired calcium handling, decreased antioxidant pathway activity, and increased reactive oxygen species production. Taken together, our data indicate that hiPSC-CMs are a suitable platform to identify and characterize the genetic basis and molecular mechanisms of DIC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Heart Failure/chemically induced , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Adult , Aged , Antibiotics, Antineoplastic/adverse effects , Calcium/metabolism , Cardiotoxicity/genetics , Cell Survival/drug effects , DNA Damage/drug effects , Disease Susceptibility , Doxorubicin/adverse effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Heart Failure/genetics , Humans , Induced Pluripotent Stem Cells , Membrane Potential, Mitochondrial/drug effects , Middle Aged , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Transcriptome
5.
Curr Protoc Hum Genet ; 87: 21.3.1-21.3.15, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26439715

ABSTRACT

Since the first discovery that human pluripotent stem cells (hPS cells) can differentiate to cardiomyocytes, efforts have been made to optimize the conditions under which this process occurs. One of the most effective methodologies to optimize this process is reductionist simplification of the medium formula, which eliminates complex animal-derived components to help reveal the precise underlying mechanisms. Here we describe our latest, cost-effective and efficient methodology for the culture of hPS cells in the pluripotent state using a modified variant of chemically defined E8 medium. We provide exact guidelines for cell handling under these conditions, including non-enzymatic EDTA passaging, which have been optimized for subsequent cardiomyocyte differentiation. We describe in depth the latest version of our monolayer chemically defined small molecule differentiation protocol, including metabolic selection-based cardiomyocyte purification and the addition of triiodothyronine to enhance cardiomyocyte maturation. Finally, we describe a method for the dissociation of hPS cell-derived cardiomyocytes, cryopreservation, and thawing.


Subject(s)
Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Cell Culture Techniques , Flow Cytometry , Humans , Immunophenotyping/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Phenotype , Pluripotent Stem Cells/metabolism
6.
J Card Fail ; 21(9): 702-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25908019

ABSTRACT

BACKGROUND: The objective of the present study was to investigate whether mild cognitive deficits are present in patients with heart failure (HF) despite absence of any known cognitive disorder. METHODS AND RESULTS: A well defined group of patients (n = 40) with heart failure completed a cognitive screening check list, a depression screening questionnaire, and a battery consisting of neuropsychological tests assessing 5 different cognitive domains: speed/attention, episodic memory, visuospatial functions, language, and executive functions. The neuropsychological results were compared with those from a group of healthy control subjects (n = 41). The patients with HF displayed cognitive impairment compared with the control group within the domains speed and attention, episodic memory, visuospatial functions, and language. Among them, 34 HF patients (85%) could be classified with mild cognitive impairment (MCI), the majority as nonamnestic MCI, ie, with no memory impairment. CONCLUSIONS: Considering the high occurrence of mild cognitive deficits among HF patients without known cognitive disorders, closer attention should be paid to their self-care and compliance. Inadequate self-care and compliance could lead to more frequent hospitalizations. Furthermore, the HF patients may be at increased risk of dementia.


Subject(s)
Attention/physiology , Cognition/physiology , Cognitive Dysfunction/etiology , Heart Failure/complications , Aged , Aged, 80 and over , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/psychology , Executive Function , Female , Follow-Up Studies , Heart Failure/psychology , Humans , Male , Middle Aged , Neuropsychological Tests , Retrospective Studies
7.
Cardiol J ; 22(1): 31-6, 2015.
Article in English | MEDLINE | ID: mdl-24526512

ABSTRACT

BACKGROUND: Due to increasing co-morbidity associated with aging, heart failure (HF) has become more prevalent and heterogeneous in older individuals, and non-cardiovascular (CV) mortality has increased. Previously, we defined a multi-marker modality that included cystatin C (CysC), troponin T (TnT), and age. Here, we validated this multi-marker risk score by evaluating its predictions of all-cause mortality and CV mortality in an independent population of older individuals with HF and reduced ejection fraction (HFrEF). METHODS: This prospective cohort study included 124 patients, median age 73 years, that had HFrEF. We determined all-cause mortality and CV mortality at a 3-year follow-up. We compared the risk score to the N-terminal prohormone of B-type natriuretic peptide (NT-proBNP) for predicting all-cause mortality and CV mortality. RESULTS: High risk scores were associated with both all-cause mortality (HR 4.2, 95% CI 2.2-8.1, p < 0.001) and CV mortality (HR 3.6, 95% CI 1.7-8.0, p = 0.0015). Receiver operating characteristics showed similar efficacy for the risk score and NT-proBNP in predicting all-cause mortality (HR 0.74, 95% CI 0.65-0.81 vs. HR 0.74, 95% CI 0.65-0.81, p = 0.99) and CV mortality (HR 0.68, 95% CI 0.59-0.76 vs. HR 0.67, 95% CI 0.58-0.75, p = 0.95). When the risk score was added to the NT-proBNP, the continuous net reclassification improvement was 56% for predicting all-cause mortality (95% CI 18-95%, p = 0.004) and 45% for predicting CV mortality (95% CI 2-89%, p = 0.040). CONCLUSIONS: In HFrEF, a risk score that included age, TnT, and CysC showed efficacy similar to the NT-proBNP for predicting all-cause mortality and CV mortality in an older population.


Subject(s)
Decision Support Techniques , Heart Failure/mortality , Stroke Volume , Age Factors , Aged , Aged, 80 and over , Area Under Curve , Biomarkers/blood , Cause of Death , Comorbidity , Cystatin C/blood , Female , Heart Failure/blood , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Predictive Value of Tests , Prognosis , Prospective Studies , ROC Curve , Reproducibility of Results , Risk Assessment , Risk Factors , Time Factors , Troponin T/blood
8.
BMC Neurol ; 13: 122, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24053888

ABSTRACT

BACKGROUND: Despite heart failure being a substantial risk factor for stroke, few studies have evaluated the predictive value of heart dysfunction for all-cause mortality in patients with acute ischemic stroke, in particular in the elderly. The aim of this study was to investigate whether impaired heart function in elderly patients can predict all-cause mortality after acute ischemic stroke or transient ischemic attack (TIA). METHODS: A prospective long-term follow-up analysis was performed on a hospital cohort consisting of n = 132 patients with mean age 73 ± 9 years, presenting with acute ischemic stroke or transient ischemic attack, without atrial fibrillation. All patients were examined by echocardiography during the hospital stay. Data about all-cause mortality were collected at the end of the follow-up period. The mean follow-up period was 56 ± 22 months. RESULTS: In this cohort, 58% of patients with acute ischemic stroke or TIA had heart dysfunction. Survival analysis showed that heart dysfunction did not predict all-cause mortality in this cohort. Furthermore, in multivariate regression analysis age (HR 5.401, Cl 1.97-14.78, p < 0.01), smoking (HR 3.181, Cl 1.36-7.47, p < 0.01), myocardial infarction (HR 2.826, Cl 1.17-6.83, p < 0.05) were independent predictors of all-cause mortality. CONCLUSION: In this population with acute ischemic stroke or TIA and without non-valvular atrial fibrillation, impaired heart function does not seem to be a significant predictor of all-cause mortality at long-term follow-up.


Subject(s)
Heart Diseases/complications , Heart Diseases/epidemiology , Ischemic Attack, Transient , Stroke , Aged , Aged, 80 and over , C-Reactive Protein/metabolism , Cholesterol/metabolism , Cohort Studies , Creatinine/blood , Female , Glomerular Filtration Rate , Hemoglobins/metabolism , Humans , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/epidemiology , Ischemic Attack, Transient/mortality , Male , Middle Aged , Predictive Value of Tests , Statistics, Nonparametric , Stroke/complications , Stroke/epidemiology , Stroke/mortality , Survival Rate , Troponin T/metabolism
9.
Int J Cardiol ; 167(6): 2747-52, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-22805544

ABSTRACT

OBJECTIVES: Epidemiological studies of elderly heart failure (HF) patients (≥ 85 years) are limited with inconsistent findings. Our objective is to confirm and extend epidemiological study in elderly (≥ 85 years) patients using the Swedish Heart Failure Registry database. METHODS: This retrospective study included 8,347 HF patients aged ≤ 65 years and 15,889 HF patients aged ≥ 85 years. Elderly population was further divided into two subgroups: 11,412 patients were 85-90 years and 4,477 patients were >90 years. RESULTS: The ≥ 85 year group was characterized by more women, higher systolic blood pressure (SBP), lower body-mass index (BMI), more than twice as many HF with normal left ventricular ejection fraction (HFNEF), higher incidence of cardiovascular and non-cardiovascular comorbidities and less use of proven therapeutics compared with the ≤ 65 year group. Compared with the 85-90 year subgroup, the > 90 year subgroup had a decline in cardiovascular and non-cardiovascular comorbidities except renal insufficiency and anaemia which continued to increase with ageing (p<0.01). Tendency was the same regardless of gender but slightly different between systolic HF (SHF) and HFNEF. In the group with HFNEF, there were more women, higher SBP, lower N-terminal pro-B-type natriuretic peptide levels, less ischaemic heart disease, more hypertension and left bundle branch block regardless of age. Atrial fibrillation was more frequent in patients with HFNEF than with SHF in the elderly group (p<0.01). Patients with HFNEF in the > 90 year subgroup had increasing incidence of ischaemic heart disease compared to 85-90 year group (p<0.01). CONCLUSIONS: HF patients ≥ 85 years had increased cardiovascular and non-cardiovascular comorbidities but with a decline from >90 years.


Subject(s)
Databases, Factual/trends , Heart Failure/diagnosis , Heart Failure/epidemiology , Registries , Age Factors , Aged , Aged, 80 and over , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , Comorbidity , Female , Humans , Hypertension/diagnosis , Hypertension/epidemiology , Male , Middle Aged , Renal Insufficiency/diagnosis , Renal Insufficiency/epidemiology , Retrospective Studies , Sweden/epidemiology
10.
Eur J Intern Med ; 23(7): 604-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22939804

ABSTRACT

BACKGROUND: Red blood cell distribution width (RDW), a measure of anisocytosis, is a prognostic biomarker for heart failure (HF). However it is still unclear how RDW is associated with heart function and established cardiac biomarkers. METHODS AND RESULTS: In a prospective hospital cohort of 296 patients referred for echocardiography because of suspected HF, blood sampling and clinical examination were performed within 24h after echocardiography. The patients were divided into four HF groups, including one group where the HF diagnosis was uncertain (gray zone). In the patients the mean age was 70 ± 11 years, 44% with systolic HF (SHF), 18% with heart failure with normal ejection fraction (HFNEF), 17% with gray zone and 21% without HF (non-HF). RDW was higher among patients with SHF and HFNEF, compared with gray zone and non-HF patients. The distribution of different variables over the RDW quartiles showed an inverse correlation between RDW levels and LVEF and a positive correlation between RDW and NT-proBNP levels. Further analysis with stepwise multiple linear regression demonstrated that NT-proBNP levels, but not LVEF, were independently correlated with RDW. CONCLUSION: In patients referred for echocardiography because of suspected HF, RDW levels were higher in patients with SHF and HFNEF. Moreover, NT-proBNP levels were independently linked with elevated RDW.


Subject(s)
Erythrocyte Indices , Heart Failure, Systolic/diagnosis , Heart Failure/diagnosis , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Aged , Aged, 80 and over , Biomarkers , Cohort Studies , Echocardiography , Female , Heart Failure/blood , Heart Failure, Systolic/blood , Humans , Linear Models , Male , Middle Aged , Prognosis , Prospective Studies , Stroke Volume
11.
Biol Trace Elem Res ; 146(2): 230-5, 2012 May.
Article in English | MEDLINE | ID: mdl-22020772

ABSTRACT

Selenium (Se) is known to regulate tumorigenesis and immunity at the nutritional and supranutritional levels. Because the immune system provides critical defenses against cancer and the athymic, immune-deficient NU/J nude mice are known to gradually develop CD8(+) and CD4(+) T cells, we investigated whether B and T cell maturation could be modulated by dietary Se and by tumorigenesis in nude mice. Fifteen homozygous nude mice were fed a Se-deficient, Torula yeast basal diet alone (Se-) or supplemented with 0.15 (Se+) or 1.0 (Se++) mg Se/kg (as Na(2)SeO(4)) for 6 months, followed by a 7-week time course of PC-3 prostate cancer cell xenograft (2 × 10(6) cells/site, 2 sites/mouse). Here, we show that peripheral B cell levels decreased in nude mice fed the Se - or Se++ diet and the CD4(+) T cell levels increased in mice fed the Se++ diet. During the PC-3 cell tumorigenesis, dietary Se status did not affect peripheral CD4(+) or CD8(+) T cells in nude mice whereas mice fed with the Se++ diet appeared to exhibit greater peripheral CD25(+)CD4(+) T cells on day 9. Dietary Se status did not affect spleen weight in nude mice 7 weeks after the xenograft. Spleen weight was associated with frequency of peripheral CD4(+), but not CD8(+) T cells. Taken together, dietary Se at the nutritional and supranutritional levels regulates peripheral B and T cells in adult nude mice before and after xenograft with PC-3 prostate cancer cells.


Subject(s)
B-Lymphocytes/drug effects , Prostatic Neoplasms/immunology , Selenium/pharmacology , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dietary Supplements , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Organ Size/drug effects , Prostatic Neoplasms/pathology , Selenium/administration & dosage , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology , Time Factors , Transplantation, Heterologous , Tumor Burden/drug effects
12.
J Nutr Biochem ; 23(9): 1086-91, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22137259

ABSTRACT

The inhibitory effect of oral methylseleninic acid or methylselenocysteine administration on cancer cell xenograft development in nude mice is well characterized; however, less is known about the efficacy of selenate and age on selenium chemoprevention. In this study, we tested whether selenate and duration on diets would regulate prostate cancer xenograft in nude mice. Thirty-nine homozygous NU/J nude mice were fed a selenium-deficient, Torula yeast basal diet alone (Se-) or supplemented with 0.15 (Se) or 1.0 (Se+) mg selenium/kg (as Na2SeO4) for 6 months in Experiment 1 and for 4 weeks in Experiment 2, followed by a 47-day PC-3 prostate cancer cell xenograft on the designated diet. In Experiment 1, the Se- diet enhanced the initial tumor development on days 11-17, whereas the Se+ diet suppressed tumor growth on days 35-47 in adult nude mice. Tumors grown in Se- mice were loosely packed and showed increased necrosis and inflammation as compared to those in Se and Se+ mice. In Experiment 2, dietary selenium did not affect tumor development or histopathology throughout the time course. In both experiments, postmortem plasma selenium concentrations in Se and Se+ mice were comparable and were twofold greater than those in Se- mice. Taken together, dietary selenate at nutritional and supranutritional levels differentially inhibit tumor development in adult, but not young, nude mice engrafted with PC-3 prostate cancer cells.


Subject(s)
Aging , Anticarcinogenic Agents/therapeutic use , Dietary Supplements , Prostatic Neoplasms/prevention & control , Selenium Compounds/therapeutic use , Animals , Anticarcinogenic Agents/administration & dosage , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Necrosis , Prostatic Neoplasms/etiology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Random Allocation , Selenic Acid , Selenium/blood , Selenium/deficiency , Selenium Compounds/administration & dosage , Time Factors , Tumor Burden , Weight Loss , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...