Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 19: 3623-3639, 2024.
Article in English | MEDLINE | ID: mdl-38660023

ABSTRACT

Introduction: Despite improvements in chemotherapy and molecularly targeted therapies, the life expectancy of patients with advanced non-small cell lung cancer (NSCLC) remains less than 1 year. There is thus a major global need to advance new treatment strategies that are more effective for NSCLC. Drug delivery using liposomal particles has shown success at improving the biodistribution and bioavailability of chemotherapy. Nevertheless, liposomal drugs lack selectivity for the cancer cells and have a limited ability to penetrate the tumor site, which severely limits their therapeutic potential. Epidermal growth factor receptor (EGFR) is overexpressed in NSCLC tumors in about 80% of patients, thus representing a promising NSCLC-specific target for redirecting liposome-embedded chemotherapy to the tumor site. Methods: Herein, we investigated the targeting of PEGylated liposomal doxorubicin (Caelyx), a powerful off-the-shelf antitumoral liposomal drug, to EGFR as a therapeutic strategy to improve the specific delivery and intratumoral accumulation of chemotherapy in NSCLC. EGFR-targeting of Caelyx was enabled through its complexing with a polyethylene glycol (PEG)/EGFR bispecific antibody fragment. Tumor targeting and therapeutic potency of our treatment approach were investigated in vitro using a panel of NSCLC cell lines and 3D tumoroid models, and in vivo in a cell line-derived tumor xenograft model. Results: Combining Caelyx with our bispecific antibody generated uniform EGFR-targeted particles with improved binding and cytotoxic efficacy toward NSCLC cells. Effects were exclusive to cancer cells expressing EGFR, and increments in efficacy positively correlated with EGFR density on the cancer cell surface. The approach demonstrated increased penetration within 3D spheroids and was effective at targeting and suppressing the growth of NSCLC tumors in vivo while reducing drug delivery to the heart. Conclusion: EGFR targeting represents a successful approach to enhance the selectivity and therapeutic potency of liposomal chemotherapy toward NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Doxorubicin , ErbB Receptors , Lung Neoplasms , Animals , Female , Humans , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/analogs & derivatives , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice, Nude , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
2.
J Control Release ; 367: 806-820, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38341177

ABSTRACT

High-risk neuroblastoma has poor survival due to treatment failure and off-target side effects of therapy. Small molecule inhibitors have shown therapeutic efficacy at targeting oncogenic cell cycle dysregulators, such as polo-like kinase 1 (PLK1). However, their clinical success is limited by a lack of efficacy and specificity, causing off-target toxicity. Herein, we investigate a new treatment strategy whereby a bispecific antibody (BsAb) with dual recognition of methoxy polyethylene glycol (PEG) and a neuroblastoma cell-surface receptor, epidermal growth factor receptor (EGFR), is combined with a PEGylated small interfering RNA (siRNA) lipid nanoparticle, forming BsAb-nanoparticle RNA-interference complexes for targeted PLK1 inhibition against high-risk neuroblastoma. Therapeutic efficacy of this strategy was explored in neuroblastoma cell lines and a tumor xenograft model. Using ionizable lipid-based nanoparticles as a low-toxicity and clinically safe approach for siRNA delivery, we identified that their complexing with EGFR-PEG BsAb resulted in increases in cell targeting (1.2 to >4.5-fold) and PLK1 gene silencing (>2-fold) against EGFR+ high-risk neuroblastoma cells, and enhancements correlated with EGFR expression on the cells (r > 0.94). Through formulating nanoparticles with PEG-lipids ranging in diffusivity, we further identified a highly diffusible PEG-lipid which provided the most pronounced neuroblastoma cell binding, PLK1 silencing, and significantly reduced cancer growth in vitro in high-risk neuroblastoma cell cultures and in vivo in a tumor-xenograft mouse model of the disease. Together, this work provides an insight on the role of PEG-lipid diffusivity and EGFR targeting as potentially relevant variables influencing the therapeutic efficacy of siRNA nanoparticles in high-risk neuroblastoma.


Subject(s)
Nanoparticles , Neuroblastoma , Humans , Animals , Mice , RNA, Small Interfering , Protein Serine-Threonine Kinases , Cell Cycle Proteins/genetics , Polo-Like Kinase 1 , Polyethylene Glycols/chemistry , Proto-Oncogene Proteins , Cell Line, Tumor , Neuroblastoma/drug therapy , ErbB Receptors/genetics , Nanoparticles/chemistry , Cell Proliferation , Lipids/pharmacology
3.
Sci Transl Med ; 15(696): eabm1262, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37196067

ABSTRACT

High-risk childhood leukemia has a poor prognosis because of treatment failure and toxic side effects of therapy. Drug encapsulation into liposomal nanocarriers has shown clinical success at improving biodistribution and tolerability of chemotherapy. However, enhancements in drug efficacy have been limited because of a lack of selectivity of the liposomal formulations for the cancer cells. Here, we report on the generation of bispecific antibodies (BsAbs) with dual binding to a leukemic cell receptor, such as CD19, CD20, CD22, or CD38, and methoxy polyethylene glycol (PEG) for the targeted delivery of PEGylated liposomal drugs to leukemia cells. This liposome targeting system follows a "mix-and-match" principle where BsAbs were selected on the specific receptors expressed on leukemia cells. BsAbs improved the targeting and cytotoxic activity of a clinically approved and low-toxic PEGylated liposomal formulation of doxorubicin (Caelyx) toward leukemia cell lines and patient-derived samples that are immunophenotypically heterogeneous and representative of high-risk subtypes of childhood leukemia. BsAb-assisted improvements in leukemia cell targeting and cytotoxic potency of Caelyx correlated with receptor expression and were minimally detrimental in vitro and in vivo toward expansion and functionality of normal peripheral blood mononuclear cells and hematopoietic progenitors. Targeted delivery of Caelyx using BsAbs further enhanced leukemia suppression while reducing drug accumulation in the heart and kidneys and extended overall survival in patient-derived xenograft models of high-risk childhood leukemia. Our methodology using BsAbs therefore represents an attractive targeting platform to potentiate the therapeutic efficacy and safety of liposomal drugs for improved treatment of high-risk leukemia.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Leukemia , Humans , Antibodies, Bispecific/therapeutic use , Tissue Distribution , Leukocytes, Mononuclear , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antineoplastic Agents/therapeutic use , Polyethylene Glycols , Liposomes , Leukemia/drug therapy
4.
Cancer Res ; 80(19): 4129-4144, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816860

ABSTRACT

Therapeutic checkpoint antibodies blocking programmed death receptor 1/programmed death ligand 1 (PD-L1) signaling have radically improved clinical outcomes in cancer. However, the regulation of PD-L1 expression on tumor cells is still poorly understood. Here we show that intratumoral copper levels influence PD-L1 expression in cancer cells. Deep analysis of the The Cancer Genome Atlas database and tissue microarrays showed strong correlation between the major copper influx transporter copper transporter 1 (CTR-1) and PD-L1 expression across many cancers but not in corresponding normal tissues. Copper supplementation enhanced PD-L1 expression at mRNA and protein levels in cancer cells and RNA sequencing revealed that copper regulates key signaling pathways mediating PD-L1-driven cancer immune evasion. Conversely, copper chelators inhibited phosphorylation of STAT3 and EGFR and promoted ubiquitin-mediated degradation of PD-L1. Copper-chelating drugs also significantly increased the number of tumor-infiltrating CD8+ T and natural killer cells, slowed tumor growth, and improved mouse survival. Overall, this study reveals an important role for copper in regulating PD-L1 and suggests that anticancer immunotherapy might be enhanced by pharmacologically reducing intratumor copper levels. SIGNIFICANCE: These findings characterize the role of copper in modulating PD-L1 expression and contributing to cancer immune evasion, highlighting the potential for repurposing copper chelators as enhancers of antitumor immunity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4129/F1.large.jpg.


Subject(s)
B7-H1 Antigen/metabolism , Brain Neoplasms/immunology , Copper/metabolism , Neuroblastoma/immunology , Tumor Escape/physiology , Animals , B7-H1 Antigen/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Chelating Agents/pharmacology , Copper Transporter 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy/methods , Killer Cells, Natural , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred BALB C , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Triethylenephosphoramide/pharmacology , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
5.
ACS Pharmacol Transl Sci ; 3(1): 148-160, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32259094

ABSTRACT

MYCN-amplified neuroblastoma is one of the deadliest forms of childhood cancer and remains a significant clinical challenge. Direct pharmacological inhibition of MYCN is not currently achievable. One strategy could be to target the AKT/GSK3ß pathway, which directly regulates the stability of the MYCN protein. Numerous potent and isoform-specific small-molecule AKT inhibitors have been developed. However, the selection of the right drug combinations in the relevant indication will have a significant impact on AKT inhibitor clinical success. To maximally exploit the potential of AKT inhibitors, a better understanding of AKT isoform functions in cancer is crucial. Here using RNAi to downregulate specific AKT isoforms, we demonstrated that loss of total AKT activity rather than isoform-specific expression was necessary to decrease MYCN expression and cause a significant decrease in neuroblastoma cell proliferation. Consistent with these observations, isoform-specific pharmacological inhibition of AKT was substantially less effective than pan-AKT inhibition in combination with cytotoxic drugs in MYCN-amplified neuroblastoma. The allosteric pan-AKT inhibitor perifosine had promising in vitro and in vivo activity in combination with conventional cytotoxic drugs in MYCN-amplified neuroblastoma cells. Our results demonstrated that perifosine drug combination was able to induce apoptosis and downregulate ABC transporter expression. Collectively, this study shows that selecting pan-AKT inhibitors rather than isoform-specific drugs to synergize with first-line chemotherapy treatment should be considered for clinical trials for aggressive neuroblastoma and, potentially, other MYCN -driven cancers.

6.
Theranostics ; 8(20): 5645-5659, 2018.
Article in English | MEDLINE | ID: mdl-30555570

ABSTRACT

Given the strong clinical evidence that copper levels are significantly elevated in a wide spectrum of tumors, copper homeostasis is considered as an emerging target for anticancer drug design. Monitoring copper levels in vivo is therefore of paramount importance when assessing the efficacy of copper-targeting drugs. Herein, we investigated the activity of the copper-targeting compound Dextran-Catechin by developing a [64Cu]CuCl2 PET imaging protocol to monitor its effect on copper homeostasis in tumors. Methods: Protein expression of copper transporter 1 (CTR1) in tissue microarrays representing 90 neuroblastoma patient tumors was assessed by immunohistochemistry. Western blotting analysis was used to study the effect of Dextran-Catechin on the expression of CTR1 in neuroblastoma cell lines and in tumors. A preclinical human neuroblastoma xenograft model was used to study anticancer activity of Dextran-Catechin in vivo and its effect on tumor copper homeostasis. PET imaging with [64Cu]CuCl2 was performed in such preclinical neuroblastoma model to monitor alteration of copper levels in tumors during treatment. Results: CTR1 protein was found to be highly expressed in patient neuroblastoma tumors by immunohistochemistry. Treatment of neuroblastoma cell lines with Dextran-Catechin resulted in decreased levels of glutathione and in downregulation of CTR1 expression, which caused a significant decrease of intracellular copper. No changes in CTR1 expression was observed in normal human astrocytes after Dextran-Catechin treatment. In vivo studies and PET imaging analysis using the neuroblastoma preclinical model revealed elevated [64Cu]CuCl2 retention in the tumor mass. Following treatment with Dextran-Catechin, there was a significant reduction in radioactive uptake, as well as reduced tumor growth. Ex vivo analysis of tumors collected from Dextran-Catechin treated mice confirmed the reduced levels of CTR1. Interestingly, copper levels in blood were not affected by treatment, demonstrating potential tumor specificity of Dextran-Catechin activity. Conclusion: Dextran-Catechin mediates its activity by lowering CTR1 and intracellular copper levels in tumors. This finding further reveals a potential therapeutic strategy for targeting copper-dependent cancers and presents a novel PET imaging method to assess patient response to copper-targeting anticancer treatments.


Subject(s)
Positron-Emission Tomography/methods , Animals , Catechin , Cation Transport Proteins , Cell Line, Tumor , Copper , Copper Transporter 1 , Dextrans , Female , Homeostasis , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred BALB C , Molecular Imaging , Neuroblastoma , Positron Emission Tomography Computed Tomography , Tissue Array Analysis
7.
Nat Commun ; 9(1): 2288, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895867

ABSTRACT

For many normal and aberrant cell behaviours, it is important to understand the origin of cellular heterogeneity. Although powerful methods for studying cell heterogeneity have emerged, they are more suitable for common rather than rare cells. Exploring the heterogeneity of rare single cells is challenging because these rare cells must be first pre-concentrated and undergo analysis prior to classification and expansion. Here, a versatile capture & release platform consisting of an antibody-modified and electrochemically cleavable semiconducting silicon surface for release of individual cells of interest is presented. The captured cells can be interrogated microscopically and tested for drug responsiveness prior to release and recovery. The capture & release strategy was applied to identify rare tumour cells from whole blood, monitor the uptake of, and response to, doxorubicin and subsequently select cells for single-cell gene expression based on their response to the doxorubicin.


Subject(s)
Electrochemical Techniques , Photochemistry/methods , Single-Cell Analysis , Animals , Cell Survival , Doxorubicin/chemistry , Female , HeLa Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , Silicon/chemistry
8.
Sci Rep ; 7(1): 7638, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28794411

ABSTRACT

Formation of blood vessels, or angiogenesis, is crucial to cancer progression. Thus, inhibiting angiogenesis can limit the growth and spread of tumors. The natural polyphenol catechin has moderate anti-tumor activity and interacts with copper, which is essential for angiogenesis. Catechin is easily metabolized in the body and this limits its clinical application. We have recently shown that conjugation of catechin with dextran (Dextran-Catechin) improves its serum stability, and exhibits potent anti-tumor activity against neuroblastoma by targeting copper homeostasis. Herein, we investigated the antiangiogenic activity of Dextran-Catechin and its mechanism. We found that Dextran-Catechin displayed potent antiangiogenic activity in vitro and in vivo. We demonstrated Dextran-Catechin generates reactive oxygen species which in turns disrupts copper homeostasis by depleting the copper importer CTR-1 and copper trafficking ATOX-1 protein. Mechanistically, we showed that disrupting copper homeostasis by knockdown of either CTR-1 or ATOX-1 protein can inhibit angiogenesis in endothelial cells. This data strongly suggests the Dextran-Catechin potent antiangiogenic activity is mediated by disrupting copper homeostasis. Thus, compounds such as Dextran-Catechin that affects both tumor growth and angiogenesis could lead the way for development of new drugs against high copper levels tumors.


Subject(s)
Antineoplastic Agents/metabolism , Catechin/metabolism , Copper/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Homeostasis/drug effects , Neovascularization, Pathologic , Animals , Antineoplastic Agents/administration & dosage , Catechin/administration & dosage , Cation Transport Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Copper Transport Proteins , Copper Transporter 1 , Dextrans/administration & dosage , Dextrans/metabolism , Disease Models, Animal , Heterografts , Humans , Metallochaperones/metabolism , Molecular Chaperones , Neoplasm Transplantation , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Reactive Oxygen Species/metabolism , Trace Elements/metabolism , Treatment Outcome
9.
Oncotarget ; 7(30): 47479-47493, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27374085

ABSTRACT

Neuroblastoma is frequently diagnosed at advanced stage disease and treatment includes high dose chemotherapy and surgery. Despite the use of aggressive therapy survival rates are poor and children that survive their disease experience long term side effects from their treatment, highlighting the need for effective and less toxic therapies. Catechin is a natural polyphenol with anti-cancer properties and limited side effects, however its mechanism of action is unknown. Here we report that Dextran-Catechin, a conjugated form of catechin that increases serum stability, is preferentially and markedly active against neuroblastoma cells having high levels of intracellular copper, without affecting non-malignant cells. Copper transporter 1 (CTR1) is the main transporter of copper in mammalian cells and it is upregulated in neuroblastoma. Functional studies showed that depletion of CTR1 expression reduced intracellular copper levels and led to a decrease in neuroblastoma cell sensitivity to Dextran-Catechin, implicating copper in the activity of this compound. Mechanistically, Dextran-Catechin was found to react with copper, inducing oxidative stress and decreasing glutathione levels, an intracellular antioxidant and regulator of copper homeostasis. In vivo, Dextran-Catechin significantly attenuated tumour growth in human xenograft and syngeneic models of neuroblastoma. Thus, Dextran-Catechin targets copper, inhibits tumour growth, and may be valuable in the treatment of aggressive neuroblastoma and other cancers dependent on copper for their growth.


Subject(s)
Antineoplastic Agents/pharmacology , Catechin/pharmacology , Copper/metabolism , Dextrans/pharmacology , Neuroblastoma/drug therapy , Animals , Cation Transport Proteins/physiology , Cell Survival/drug effects , Copper Transporter 1 , Female , Humans , Mice , Mice, Inbred BALB C , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oxidative Stress/drug effects
10.
Oncotarget ; 6(14): 12020-34, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25557168

ABSTRACT

Non-small cell lung cancer (NSCLC) remains the most common cause of cancer death worldwide due its resistance to chemotherapy and aggressive tumor growth. Polo-like kinase 1 (PLK1) is a serine-threonine protein kinase which is overexpressed in cancer cells, and plays a major role in regulating tumor growth. A number of PLK1 inhibitors are in clinical trial; however, poor tumor bioavailability and off-target effects limit their efficacy. Short-interfering-RNA (siRNA) holds promise as a class of therapeutics, which can selectively silence disease-causing genes. However, siRNA cannot enter cells without a delivery vehicle. Herein, we investigated whether RNAi-interfering nanoparticles could deliver siRNA to NSCLC cells and silence PLK1 expression in vitro and in vivo. iNOP-7 was non-toxic, and delivered siRNA with high efficiency to NSCLC cells. iNOP-7-PLK1 siRNA silenced PLK1 expression and reduced NSCLC growth in vitro. Notably, iNOP-7 delivered siRNA to orthotopic lung tumors in mice, and administration of iNOP-7-PLK1 siRNA reduced lung tumor burden. These novel data show that iNOP-7 can deliver siRNA against PLK1 to NSCLC cells, and decrease cell proliferation both in vitro and in vivo. iNOP-7-PLK1 siRNA may provide a novel therapeutic strategy for the treatment of NSCLC as well as other cancers which aberrantly express this gene.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Cell Cycle Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA Interference/immunology , RNA, Small Interfering/genetics , Animals , Cell Proliferation , Humans , Mice , Nanoparticles , Transfection , Polo-Like Kinase 1
11.
Nanoscale ; 3(3): 919-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173998

ABSTRACT

We demonstrate that oral delivery of self-assembled nanostructured nanoparticles consisting of 5-fluorouracil (5-FU) lipid prodrugs results in a highly effective, target-activated, chemotherapeutic agent, and offers significantly enhanced efficacy over a commercially available alternative that does not self-assemble. The lipid prodrug nanoparticles have been found to significantly slow the growth of a highly aggressive mouse 4T1 breast tumour, and essentially halt the growth of a human MDA-MB-231 breast tumour in mouse xenografts. Systemic toxicity is avoided as prodrug activation requires a three-step, enzymatic conversion to 5-FU, with the third step occurring preferentially at the tumour site. Additionally, differences in the lipid prodrug chemical structure and internal nanostructure of the nanoparticle dictate the enzymatic conversion rate and can be used to control sustained release profiles. Thus, we have developed novel oral nanomedicines that combine sustained release properties with target-selective activation.


Subject(s)
Breast Neoplasms/drug therapy , Fluorouracil/administration & dosage , Lipids/chemistry , Nanocapsules/administration & dosage , Administration, Oral , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Crystallization/methods , Drug Compounding/methods , Fluorouracil/chemistry , Humans , Mice , Mice, Inbred BALB C , Nanocapsules/chemistry , Nanomedicine/methods , Prodrugs , Treatment Outcome
12.
Physiol Biochem Zool ; 82(3): 270-9, 2009.
Article in English | MEDLINE | ID: mdl-19331583

ABSTRACT

Phenotypic plasticity of the gastrointestinal tract is crucial for optimal food processing and nutrient balance in many vertebrate species. For mammalian herbivores, gut plasticity is typically correlated with the fiber content of forage; however, we show here that other factors such as ingesta particle size may effect profound phenotypic plasticity of the fermentative hindgut in a medium-sized (10-kg body mass) marsupial herbivore, the red-necked wallaby (Macropus rufogriseus). When dietary fiber contents were comparable, red-necked wallabies that were fed a finely ground, pelleted hay for 60-72 d had hindguts that were some 28% heavier (empty wet mass) than those fed unchopped hay. The hindguts of pellet-fed wallabies contained more wet ingesta, which was also of a finer particle size, than those fed hay, indicating some separation of large- and small-particle fermentation between the foregut and the hindgut, respectively. Such a digestive strategy would benefit animals by allowing fermentation of a range of ingesta particle sizes that are expected for free-ranging animals faced with a spectrum of diet types and qualities. The heavier hindgut of pellet-fed wallabies was correlated with increased concentrations of short-chain fatty acids (SCFAs) in the fermentative hindgut (cecum and proximal colon) and particularly with increases in the molar proportions of n-butyric acid. The mechanisms facilitating gut plasticity in herbivorous mammals are uncertain, but we suggest that manipulating ingesta particle size rather than dietary fiber could provide a useful tool for evaluating causal explanations. In particular, altering ingesta particle size could help to distinguish possible direct processes (e.g., the favoring of smaller intestinal microbes and production of specific SCFAs) from indirect affects of feed structure (e.g., muscular hypertrophy to compensate for increased intakes and digesta bulk or the fermentation of mucus secreted to promote the flow of viscous, fine-particle material).


Subject(s)
Adaptation, Physiological , Animal Feed , Dietary Fiber , Gastrointestinal Tract/physiology , Macropodidae/physiology , Animals , Energy Metabolism , Female , Gastrointestinal Tract/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL
...