23 results on '"Natalie O. Karpinich"'
Search Results
2. Data from A Phase I/II Open-Label Study of Molibresib for the Treatment of Relapsed/Refractory Hematologic Malignancies
- Author
-
Michael Dickinson, Brandon E. Kremer, Arindam Dhar, Thierry Horner, Shawn W. Foley, Michael T. McCabe, Natalie O. Karpinich, Geraldine Ferron-Brady, Evi Bakirtzi, Olena Barbash, Anu Shilpa Krishnatry, Anastasia Wyce, Paolo Gallipoli, Ludovica Marando, Faisal Basheer, Regina García Delgado, Paul Yeh, Natalia Tovar, Maria-Victoria Mateos, Manali Kamdar, Jacob L. Glass, Gareth J. Morgan, Faith E. Davies, Daniel A. Pollyea, Dan T. Vogl, Aristeidis Chaidos, Adrian Alegre, Anastasios Karadimitris, Brian J.P. Huntly, Gautam Borthakur, and Mark A. Dawson
- Abstract
Purpose:Molibresib is a selective, small molecule inhibitor of the bromodomain and extra-terminal (BET) protein family. This was an open-label, two-part, Phase I/II study investigating molibresib monotherapy for the treatment of hematological malignancies (NCT01943851).Patients and Methods:Part 1 (dose escalation) determined the recommended Phase 2 dose (RP2D) of molibresib in patients with acute myeloid leukemia (AML), Non–Hodgkin lymphoma (NHL), or multiple myeloma. Part 2 (dose expansion) investigated the safety and efficacy of molibresib at the RP2D in patients with relapsed/refractory myelodysplastic syndrome (MDS; as well as AML evolved from antecedent MDS) or cutaneous T-cell lymphoma (CTCL). The primary endpoint in Part 1 was safety and the primary endpoint in Part 2 was objective response rate (ORR).Results:There were 111 patients enrolled (87 in Part 1, 24 in Part 2). Molibresib RP2Ds of 75 mg daily (for MDS) and 60 mg daily (for CTCL) were selected. Most common Grade 3+ adverse events included thrombocytopenia (37%), anemia (15%), and febrile neutropenia (15%). Six patients achieved complete responses [3 in Part 1 (2 AML, 1 NHL), 3 in Part 2 (MDS)], and 7 patients achieved partial responses [6 in Part 1 (4 AML, 2 NHL), 1 in Part 2 (MDS)]. The ORRs for Part 1, Part 2, and the total study population were 10% [95% confidence interval (CI), 4.8–18.7], 25% (95% CI, 7.3–52.4), and 13% (95% CI, 6.9–20.6), respectively.Conclusions:While antitumor activity was observed with molibresib, use was limited by gastrointestinal and thrombocytopenia toxicities. Investigations of molibresib as part of combination regimens may be warranted.
- Published
- 2023
- Full Text
- View/download PDF
3. Phase I trials of the lysine-specific demethylase 1 inhibitor, GSK2879552, as mono- and combination-therapy in relapsed/refractory acute myeloid leukemia or high-risk myelodysplastic syndromes
- Author
-
Gail J. Roboz, Karen Yee, Amit Verma, Gautam Borthakur, Adolfo de la Fuente Burguera, Guillermo Sanz, Helai P. Mohammad, Ryan G. Kruger, Natalie O. Karpinich, Geraldine Ferron-Brady, Andre Acusta, Heather Del Buono, Therese Collingwood, Marc Ballas, Arindam Dhar, and Andrew H. Wei
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
- Full Text
- View/download PDF
4. A phase I/II open-label study of molibresib for the treatment of relapsed/refractory hematologic malignancies
- Author
-
Mark A. Dawson, Gautam Borthakur, Brian J.P. Huntly, Anastasios Karadimitris, Adrian Alegre, Aristeidis Chaidos, Dan T. Vogl, Daniel A. Pollyea, Faith E. Davies, Gareth J. Morgan, Jacob L. Glass, Manali Kamdar, Maria-Victoria Mateos, Natalia Tovar, Paul Yeh, Regina García Delgado, Faisal Basheer, Ludovica Marando, Paolo Gallipoli, Anastasia Wyce, Anu Shilpa Krishnatry, Olena Barbash, Evi Bakirtzi, Geraldine Ferron-Brady, Natalie O. Karpinich, Michael T. McCabe, Shawn W. Foley, Thierry Horner, Arindam Dhar, Brandon E. Kremer, and Michael Dickinson
- Subjects
Cancer Research ,Oncology - Abstract
Purpose:Molibresib is a selective, small molecule inhibitor of the bromodomain and extra-terminal (BET) protein family. This was an open-label, two-part, Phase I/II study investigating molibresib monotherapy for the treatment of hematological malignancies (NCT01943851).Patients and Methods:Part 1 (dose escalation) determined the recommended Phase 2 dose (RP2D) of molibresib in patients with acute myeloid leukemia (AML), Non–Hodgkin lymphoma (NHL), or multiple myeloma. Part 2 (dose expansion) investigated the safety and efficacy of molibresib at the RP2D in patients with relapsed/refractory myelodysplastic syndrome (MDS; as well as AML evolved from antecedent MDS) or cutaneous T-cell lymphoma (CTCL). The primary endpoint in Part 1 was safety and the primary endpoint in Part 2 was objective response rate (ORR).Results:There were 111 patients enrolled (87 in Part 1, 24 in Part 2). Molibresib RP2Ds of 75 mg daily (for MDS) and 60 mg daily (for CTCL) were selected. Most common Grade 3+ adverse events included thrombocytopenia (37%), anemia (15%), and febrile neutropenia (15%). Six patients achieved complete responses [3 in Part 1 (2 AML, 1 NHL), 3 in Part 2 (MDS)], and 7 patients achieved partial responses [6 in Part 1 (4 AML, 2 NHL), 1 in Part 2 (MDS)]. The ORRs for Part 1, Part 2, and the total study population were 10% [95% confidence interval (CI), 4.8–18.7], 25% (95% CI, 7.3–52.4), and 13% (95% CI, 6.9–20.6), respectively.Conclusions:While antitumor activity was observed with molibresib, use was limited by gastrointestinal and thrombocytopenia toxicities. Investigations of molibresib as part of combination regimens may be warranted.
- Published
- 2022
5. Gap Junction Coupling Is Required for Tumor Cell Migration Through Lymphatic Endothelium
- Author
-
Kathleen M. Caron and Natalie O. Karpinich
- Subjects
Cell signaling ,Pathology ,medicine.medical_specialty ,government.form_of_government ,Cell Communication ,Biology ,Article ,Adrenomedullin ,chemistry.chemical_compound ,Cell Movement ,Tumor Cells, Cultured ,Lymphatic vessel ,medicine ,Humans ,Cells, Cultured ,beta Catenin ,Lucifer yellow ,Gap junction ,Gap Junctions ,Biological Transport ,Neoplastic Cells, Circulating ,Coculture Techniques ,Cell biology ,Endothelial stem cell ,Lymphatic Endothelium ,medicine.anatomical_structure ,Lymphatic system ,chemistry ,Lymphatic Metastasis ,government ,Endothelium, Lymphatic ,Cardiology and Cardiovascular Medicine - Abstract
Objective— The lymphatic vasculature is a well-established conduit for metastasis, but the mechanisms by which tumor cells interact with lymphatic endothelial cells (LECs) to facilitate escape remain poorly understood. Elevated levels of the lymphangiogenic peptide adrenomedullin are found in many tumors, and we previously characterized that its expression is necessary for lymphatic vessel growth within both tumors and sentinel lymph nodes and for distant metastasis. Approach and Results— This study used a tumor cell-LEC coculture system to identify a series of adrenomedullin-induced events that facilitated transendothelial migration of the tumor cells through a lymphatic monolayer. High levels of adrenomedullin expression enhanced adhesion of tumor cells to LECs, and further analysis revealed that adrenomedullin promoted gap junction coupling between LECs as evidenced by spread of Lucifer yellow dye. Adrenomedullin also enhanced heterocellular gap junction coupling as demonstrated by Calcein dye transfer from tumor cells into LECs. This connexin-mediated gap junction intercellular communication was necessary for tumor cells to undergo transendothelial migration because pharmacological blockade of this heterocellular communication prevented the ability of tumor cells to transmigrate through the lymphatic monolayer. In addition, treatment of LECs with adrenomedullin caused nuclear translocation of β-catenin, a component of endothelial cell junctions, causing an increase in transcription of the downstream target gene C-MYC . Importantly, blockade of gap junction intercellular communication prevented β-catenin nuclear translocation. Conclusions— Our findings indicate that maintenance of cell–cell communication is necessary to facilitate a cascade of events that lead to tumor cell migration through the lymphatic endothelium.
- Published
- 2015
- Full Text
- View/download PDF
6. Midregional pro-adrenomedullin plasma concentrations are blunted in severe preeclampsia
- Author
-
William Valdar, Kathleen M. Caron, Amy P. Murtha, Robert W. Corty, Brooke C. Matson, Natalie O. Karpinich, and Chad A. Grotegut
- Subjects
Adult ,Placental growth factor ,medicine.medical_specialty ,Complications of pregnancy ,Article ,Preeclampsia ,Sepsis ,Adrenomedullin ,Young Adult ,Pre-Eclampsia ,Pregnancy ,Internal medicine ,medicine ,Humans ,Protein Precursors ,business.industry ,Obstetrics and Gynecology ,Endoglin ,medicine.disease ,Endocrinology ,Reproductive Medicine ,Case-Control Studies ,Biomarker (medicine) ,Female ,business ,Biomarkers ,Developmental Biology - Abstract
Levels of the peptide hormone adrenomedullin (AM) are elevated during normal pregnancy, but whether this differs during complications of pregnancy remains unresolved. AM can be quantified by measuring its preprohormone byproduct, midregional pro-adrenomedullin (MR-proADM). MR-pro ADM has shown prognostic value as a biomarker of heart failure, sepsis, and community-acquired pneumonia. Given the relevance of AM to pregnancy, we tested the hypothesis that MR-proADM provides a biomarker for preeclampsia. We find that MR-proADM plasma concentrations are blunted in severe preeclampsia and that MR-proADM is similarly effective as established biomarkers endoglin and placental growth factor at discriminating patients with severe preeclampsia from controls.
- Published
- 2014
- Full Text
- View/download PDF
7. Decoy Receptor CXCR7 Modulates Adrenomedullin-Mediated Cardiac and Lymphatic Vascular Development
- Author
-
Helen H. Willcockson, Klara R. Klein, Erich J. Kushner, William P. Dunworth, Scott T. Espenschied, Kathleen M. Caron, Natalie O. Karpinich, Victoria L. Bautch, and Samantha L. Hoopes
- Subjects
Male ,medicine.medical_specialty ,Biology ,Ligands ,General Biochemistry, Genetics and Molecular Biology ,Adrenomedullin ,Mice ,Cell Movement ,Internal medicine ,medicine ,Animals ,Humans ,Decoy receptors ,Receptor ,Molecular Biology ,Cell Proliferation ,Lymphatic Vessels ,Oligonucleotide Array Sequence Analysis ,G protein-coupled receptor ,Mice, Knockout ,Receptors, CXCR ,Regulation of gene expression ,Muscle Cells ,HEK 293 cells ,Gene Expression Regulation, Developmental ,Heart ,Cell Biology ,Cell biology ,HEK293 Cells ,Phenotype ,Endocrinology ,Female ,Signal transduction ,Decoy ,Signal Transduction ,Developmental Biology - Abstract
SummaryAtypical 7-transmembrane receptors, often called decoy receptors, act promiscuously as molecular sinks to regulate ligand bioavailability and consequently temper the signaling of canonical G protein-coupled receptor (GPCR) pathways. Loss of mammalian CXCR7, the most recently described decoy receptor, results in postnatal lethality due to aberrant cardiac development and myocyte hyperplasia. Here, we provide the molecular underpinning for this proliferative phenotype by demonstrating that the dosage and signaling of adrenomedullin (Adm, gene; AM, protein)—a mitogenic peptide hormone required for normal cardiovascular development—is tightly controlled by CXCR7. To this end, Cxcr7−/− mice exhibit gain-of-function cardiac and lymphatic vascular phenotypes that can be reversed upon genetic depletion of adrenomedullin ligand. In addition to identifying a biological ligand accountable for the phenotypes of Cxcr7−/− mice, these results reveal a previously underappreciated role for decoy receptors as molecular rheostats in controlling the timing and extent of GPCR-mediated cardiac and vascular development.
- Published
- 2014
- Full Text
- View/download PDF
8. Adrenomedullin Function in Vascular Endothelial Cells: Insights from Genetic Mouse Models
- Author
-
Kathleen M. Caron, Daniel O. Kechele, Patricia M. Lenhart, Samantha L. Hoopes, and Natalie O. Karpinich
- Subjects
CLR ,business.industry ,Angiogenesis ,mouse model ,RAMPs ,Disease ,Bioinformatics ,Article ,Lymphangiogenesis ,Cell biology ,lymphangiogenesis ,Adrenomedullin ,angiogenesis ,Tumor progression ,endothelial ,Internal Medicine ,Medicine ,permeability ,business ,hormones, hormone substitutes, and hormone antagonists ,Function (biology) - Abstract
Adrenomedullin is a highly conserved peptide implicated in a variety of physiological processes ranging from pregnancy and embryonic development to tumor progression. This review highlights past and present studies that have contributed to our current appreciation of the important roles adrenomedullin plays in both normal and disease conditions. We provide a particular emphasis on the functions of adrenomedullin in vascular endothelial cells and how experimental approaches in genetic mouse models have helped to drive the field forward.
- Published
- 2011
- Full Text
- View/download PDF
9. ERK-1 MAP kinase prevents TNF-induced apoptosis through bad phosphorylation and inhibition of Bax translocation in HeLa Cells
- Author
-
John L. Farber, Natalie O. Karpinich, Valentina Paradisi, Matteo Antonio Russo, Massimo Fini, Michele Aventaggiato, Marco Tafani, Valentina Reali, Bruna Pucci, Manuela Indelicato, and Laura Pellegrini
- Subjects
MAPK/ERK pathway ,Programmed cell death ,TNF ,Apoptosis ,Biochemistry ,HeLa ,chemistry.chemical_compound ,Bcl-2-associated X protein ,Erk-1 ,Humans ,Cycloheximide ,Phosphorylation ,Molecular Biology ,Anisomycin ,bcl-2-Associated X Protein ,Caspase 8 ,Mitogen-Activated Protein Kinase 3 ,Bad phosphorylation ,Bax ,Amino Acid Substitution ,Gene Targeting ,HeLa Cells ,JNK Mitogen-Activated Protein Kinases ,Mitochondria ,Protein Transport ,Signal Transduction ,Tumor Necrosis Factors ,bcl-Associated Death Protein ,Cell Biology ,biology ,Kinase ,biology.organism_classification ,Molecular biology ,Cell biology ,chemistry ,biology.protein ,Tumor necrosis factor alpha - Abstract
Extracellular signal-regulated kinase (ERK) 1/2 signaling is involved in tumor cell survival through the regulation of Bcl-2 family members. To explore this further and to demonstrate the central role of the mitochondria in the ERK1/2 pathway we used the HeLa cellular model where apoptosis was induced by tumor necrosis factor (TNF) and cycloheximide (CHX). We show that HeLa cells overexpressing ERK-1 displayed resistance to TNF and CHX. HeLa cells overexpressing a kinase-deficient form of ERK-1 (K71R) were more sensitive to TNF and CHX. In the ERK-1 cells, Bad was phosphorylated during TNF + CHX treatment. In the HeLa wt cells and in the K71R clones TNF and CHX decreased Bad phosphorylation. ERK-1 cells treated with TNF and CHX did not release cytochrome c from the mitochondria. By contrast, HeLa wt and K71R clones released cytochrome c. Bax did not translocate to the mitochondria in ERK-1 cells treated with TNF + CHX. Conversely, HeLa wt and K71R clones accumulated Bax in the mitochondria. In the HeLa wt cells and in both ERK-1 transfectants Bid was cleaved and accumulated in the mitochondria. The caspase-8 inhibitor IETD-FMK and the mitochondrial membrane permeabilization inhibitor bongkrekic acid (BK), partially prevented cell death by TNF + CHX. Anisomycin, a c-Jun N-terminal kinases activator, increased TNF-killing. The ERK-1 cells were resistant to TNF and anisomycin, whereas K71R clones resulted more sensitive. Our study demonstrates that in HeLa cells the ERK-1 kinase prevents TNF + CHX apoptosis by regulating the intrinsic mitochondrial pathway through different mechanisms. Inhibition of the intrinsic pathway is sufficient to almost completely prevent cell death.
- Published
- 2009
- Full Text
- View/download PDF
10. Regulation of Intracellular pH Mediates Bax Activation in HeLa Cells Treated with Staurosporine or Tumor Necrosis Factor-α
- Author
-
Matteo Antonio Russo, Marco Tafani, John L. Farber, Joshua A. Cohn, Ronald J. Rothman, and Natalie O. Karpinich
- Subjects
Programmed cell death ,Time Factors ,Cell Survival ,Truncated BID ,Intracellular pH ,Blotting, Western ,Apoptosis ,Cytochrome c Group ,Chromosomal translocation ,Biochemistry ,Cytosol ,Chloride Channels ,Furosemide ,Proto-Oncogene Proteins ,medicine ,Humans ,Staurosporine ,Cycloheximide ,Enzyme Inhibitors ,Diuretics ,Molecular Biology ,bcl-2-Associated X Protein ,Protein Synthesis Inhibitors ,Caspase 8 ,biology ,Tumor Necrosis Factor-alpha ,Cytochrome c ,Cell Biology ,Hydrogen-Ion Concentration ,Caspase Inhibitors ,Molecular biology ,Caspase 9 ,Mitochondria ,Cell biology ,Protein Transport ,Cell killing ,Proto-Oncogene Proteins c-bcl-2 ,Calibration ,biology.protein ,Carrier Proteins ,Oligopeptides ,BH3 Interacting Domain Death Agonist Protein ,HeLa Cells ,Protein Binding ,medicine.drug - Abstract
Induction of apoptosis in HeLa cells with staurosporine produced a rise in the intracellular pH (pH(i)). Intracellular alkalinization was accompanied by translocation of Bax to the mitochondria, cytochrome c release, and cell death. The chloride channel inhibitor furosemide prevented intracellular alkalinization, Bax translocation, cytochrome c release, and cell death. Translocation of full-length Bid to the mitochondria was also prevented by furosemide. The cleavage product of Bid degradation (truncated Bid, tBid) was not detectable in the mitochondria. Its accumulation in the cytosol was prevented by furosemide. Apoptosis induced by tumor necrosis factor-alpha (TNF) lowered pH(i), an effect also accompanied by Bax translocation, cytochrome c release, and cell killing. Furosemide prevented all of these events. TNF induced a depletion of full-length Bid from the mitochondria and the cytosol but induced an accumulation of mitochondrial tBid. Furosemide only delayed full-length Bid depletion and tBid accumulation. The caspase 8 inhibitor IETD did not prevent the translocation of Bax. Although IETD did inhibit the cleavage of Bid and the accumulation of tBid, cell killing was reduced only slightly. It is concluded that with either staurosporine or TNF a furosemide-sensitive change in pH(i) is linked to Bax translocation, cytochrome c release, and cell killing. With TNF Bax translocation occurs as Bid is depleted and can be dissociated from the accumulation of tBid. With staurosporine a role for full-length Bid in Bax translocation cannot be excluded but is not necessary as evidenced by the data with TNF.
- Published
- 2002
- Full Text
- View/download PDF
11. The Course of Etoposide-induced Apoptosis from Damage to DNA and p53 Activation to Mitochondrial Release of Cytochromec
- Author
-
Marco Tafani, Matteo Antonio Russo, Ronald J. Rothman, John L. Farber, and Natalie O. Karpinich
- Subjects
Time Factors ,Cytochrome ,Ubiquinone ,Apoptosis ,Biochemistry ,Wortmannin ,Mice ,chemistry.chemical_compound ,Cytosol ,Furosemide ,Cyclosporin a ,Cycloheximide ,Enzyme Inhibitors ,Phosphorylation ,Diuretics ,Cells, Cultured ,Etoposide ,bcl-2-Associated X Protein ,Protein Synthesis Inhibitors ,biology ,Cytochrome c ,Mitochondria ,Up-Regulation ,Cell biology ,Protein Transport ,Proto-Oncogene Proteins c-bcl-2 ,Cell Survival ,Blotting, Western ,Cytochrome c Group ,Transfection ,Cell Line ,Proto-Oncogene Proteins c-myc ,Bcl-2-associated X protein ,Chloride Channels ,Proto-Oncogene Proteins ,Animals ,Molecular Biology ,Nucleic Acid Synthesis Inhibitors ,Cell Biology ,Fibroblasts ,Molecular biology ,Androstadienes ,Mitochondrial permeability transition pore ,chemistry ,biology.protein ,Tumor Suppressor Protein p53 ,DNA Damage - Abstract
Treatment of L929 fibroblasts by the topoisomerase II inhibitor etoposide killed 50% of the cells within 72 h. The cell killing was preceded by the release of cytochrome c from the mitochondria. Simultaneous treatment of the cells with wortmannin, cycloheximide, furosemide, cyclosporin A, or decylubiquinone prevented the release of cytochrome c and significantly reduced the loss of viability. Etoposide caused the phosphorylation of p53 within 6 h, an effect prevented by wortmannin, an inhibitor of DNA-dependent protein kinase (DNA-PK). The activation of p53 by etoposide resulted in the up-regulation of the pro-apoptotic protein Bax, a result that was prevented by the protein synthesis inhibitor cycloheximide. The increase in the content of Bax was followed by the translocation of this protein from the cytosol to the mitochondria, an event that was inhibited by furosemide, a chloride channel inhibitor. Stably transfected L929 fibroblasts that overexpress Akt were resistant to etoposide and did not translocate Bax to the mitochondria or release cytochrome c. Bax levels in these transfected cells were comparable with the wild-type cells. The release of cytochrome c upon translocation of Bax has been attributed to induction of the mitochondrial permeability transition (MPT). Cyclosporin A and decylubiquinone, inhibitors of MPT, prevented the release of cytochrome c without affecting Bax translocation. These data define a sequence of biochemical events that mediates the apoptosis induced by etoposide. This cascade proceeds by coupling DNA damage to p53 phosphorylation through the action of DNA-PK. The activation of p53 increases Bax synthesis. The translocation of Bax to the mitochondria induces the MPT, the event that releases cytochrome c and culminates in the death of the cells.
- Published
- 2002
- Full Text
- View/download PDF
12. Schlemm's canal: more than meets the eye, lymphatics in disguise
- Author
-
Kathleen M. Caron and Natalie O. Karpinich
- Subjects
Schlemm's canal ,Endothelium ,business.industry ,Ocular Pathology ,Cellular pathways ,Glaucoma ,Aqueous humor ,General Medicine ,medicine.disease ,Systemic circulation ,medicine.anatomical_structure ,Lymphatic system ,Commentary ,Medicine ,sense organs ,business ,Neuroscience - Abstract
Schlemm’s canal (SC) is a unique vascular structure that functions to maintain fluid homeostasis by draining aqueous humor from the eye into the systemic circulation. The endothelium lining the inner wall of SC has both blood and lymphatic vascular characteristics, thus prompting exploration of the development and regulation of this unique channel. In this issue of the JCI, back-to-back papers by Aspelund et al. and Park et al. detail the mechanisms of SC development, which includes a lymphatic reprogramming that is necessary to maintain proper function. Furthermore, both groups exploit the lymph-like qualities of this canal: they identify VEGF-C as a potential therapeutic for glaucoma and suggest that expression of PROX1, a marker of lymphatic fate, could also serve as a biosensor for monitoring SC integrity. These studies provide substantial insight into the molecular and cellular pathways that govern SC development and reveal that ocular pathology is associated with deregulation of the lymph-like characteristics of SC.
- Published
- 2014
13. Apelin signaling: new G protein-coupled receptor pathway in lymphatic vascular development
- Author
-
Natalie O. Karpinich and Kathleen M. Caron
- Subjects
biology ,Lymphatic endothelial cell migration ,Zebrafish Proteins ,biology.organism_classification ,Article ,Apelin ,Lymphangiogenesis ,Cell biology ,Thoracic Duct ,medicine.anatomical_structure ,Lymphatic system ,Immunology ,Lymphatic vessel ,medicine ,Animals ,Humans ,Signal transduction ,Chemokines ,Cardiology and Cardiovascular Medicine ,Zebrafish ,Apelin receptor ,Signal Transduction - Abstract
The identification of novel factors that orchestrate the development, growth, and function of the lymphatic vascular system is a relatively new and coveted goal in the broad field of vascular biology. With the increased incidence of lymphedema and the recent recognition of the important roles that lymphatic vessels play in cancer, obesity, and metabolic disorders, there is a strong motivation to identify factors that could serve as pharmacological targets for the therapeutic modulation of lymphatic vessel growth and function.1 Because G protein–coupled receptors constitute the largest proportion of targets for prescribed pharmaceuticals, there is intense interest in revealing novel receptor pathways that could be targeted with highly specific agonists and antagonists.2 In this issue of Arteriosclerosis, Thrombosis, and Vascular Biology , Kim et al3 discover a new player in lymphatic vessels: Apelin signaling—a broadly expressed and multifunctional G protein–coupled receptors pathway that is part of the broader family of adipocytokine signaling.4,5 See accompanying article on page 338 Much of our knowledge on lymphatic vessel growth has come from developmental studies, whereby the establishment of lymphatic progenitor fate, migration, proliferation, and maturation can be elegantly teased apart using genetic approaches in mice and aquatic animal model systems.6,7 Lymphatic progenitors arise from venous endothelial cells, and the process of lymphatic endothelial cell migration away from parental veins can be stereotypically visualized and quantitated in zebrafish. Therefore, identifying key growth factors that are spatiotemporally expressed during this critical point of lymphatic vessel formation provides a strong foundation for the elucidation of new players. This pattern of highly restricted expression is precisely what led Kim et al to explore the possible functions of Apelin signaling in lymphatic development. Previous studies had shown that although the expression of Apelin receptors was broad during early zebrafish development, …
- Published
- 2014
14. Characterization of class II apurinic/apyrimidinic endonuclease activities in the human malaria parasite, Plasmodium falciparum
- Author
-
Natalie O. Karpinich, Brett M. Haltiwanger, and Theodore F. Taraschi
- Subjects
chemistry.chemical_classification ,biology ,DNA repair ,Plasmodium falciparum ,Cell Biology ,Base excision repair ,biology.organism_classification ,Biochemistry ,Molecular biology ,Plasmodium ,AP endonuclease ,chemistry.chemical_compound ,Enzyme ,chemistry ,parasitic diseases ,biology.protein ,AP site ,Molecular Biology ,DNA - Abstract
We have reported that the human malaria parasite, Plasmodium falciparum, repairs apurinic/apyrimidinic (AP) sites on DNA by a long-patch base excision repair (BER) pathway. This biology is different from that in mammalian cells, which predominantly repair AP sites by a DNA-polymerase-β-dependent, one-nucleotide patch BER pathway. As a starting point for the identification and biochemical characterization of the enzymes involved in the parasite DNA BER pathway, we chose characterization of the AP endonuclease activity in a P. falciparum cell-free lysate. Evidence is provided for the presence of class II, Mg2+-dependent and independent AP endonucleases in the parasite lysate. The investigation of the processing of AP sites in Plasmodium will provide new information about long-patch BER pathways; if they are different from those in the human host they might provide a new target for anti-malarial chemotherapy.
- Published
- 1999
- Full Text
- View/download PDF
15. 305-POS
- Author
-
William Valdar, Natalie O. Karpinich, Amy P. Murtha, Brooke C. Matson, Chad A. Grotegut, Kathleen M. Caron, and Robert W. Corty
- Subjects
Placental growth factor ,medicine.medical_specialty ,Pregnancy ,business.industry ,Obstetrics and Gynecology ,Endoglin ,medicine.disease ,Gastroenterology ,Preeclampsia ,Adrenomedullin ,Endocrinology ,Internal medicine ,Internal Medicine ,medicine ,Gestation ,Biomarker (medicine) ,business ,Prospective cohort study - Abstract
Objectives Plasma levels of adrenomedullin (AM), an anti-inflammatory, vasodilatory peptide hormone, are physiologically elevated during a healthy pregnancy, but it is uncertain whether these levels are altered during complications of pregnancy like preeclampsia. A byproduct of the preprohormone of AM, midregional pro-adrenomedullin (MR-proADM), is produced in a one-to-one ratio with mature AM and can be measured as a surrogate analyte for AM. Therefore, we tested the hypothesis that MR-proADM can serve as a biomarker for preeclampsia. Methods We selected 30 women with preeclampsia and 30 normotensive controls from a prospective study that collected blood and tissue samples as patients presented to Duke University Hospital for delivery. Exclusion criteria were: mild preeclampsia ( n = 5), postpartum plasma collection ( n = 12), twin pregnancies ( n = 3), and smokers ( n = 7). Endoglin, placental growth factor (PlGF), and MR-proADM concentrations were quantified in plasma using a commercial ELISA kit or immunofluorescent assay. Endoglin, PlGF, and MR-proADM were then analyzed as biomarkers of severe preeclampsia by comparing the areas under the curve (AUC) of receiver operating characteristic (ROC) curves generated by logistic regression models of these analytes. Results MR-proADM concentrations were blunted in plasma from women with severe preeclampsia (Matson et al., 2014). In our dataset, endoglin (AUC = 0.73) and MR-proADM (AUC = 0.69) were similarly effective at classifying women with severe preeclampsia and controls, while PlGF was more informative (AUC = 0.85). Endoglin and MR-proADM together (AUC = 0.80) discriminate patients from controls better than they do individually, as do PlGF and MR-proADM together (AUC = 0.87). Conclusions MR-proADM can classify women with severe preeclampsia and controls and may provide a biomarker for severe preeclampsia. However, analysis of MR-proADM levels at earlier gestational ages is needed to determine whether MR-proADM can predict the development of preeclampsia later in pregnancy. Disclosures B. Matson: Research Support Recipient: Ferring Innovation Fellowship. N. Karpinich: None. A. Murtha: None. W. Valdar: None. C.A. Grotegut: None. K.M. Caron: Research Support Recipient; Commercial Interest: Ferring Pharmaceuticals, Inc.
- Published
- 2015
- Full Text
- View/download PDF
16. Cooperativity between MAPK and PI3K signaling activation is required for glioblastoma pathogenesis
- Author
-
C. Ryan Miller, Kristen K. White, Robert S. McNeill, Ralf S. Schmid, Sophie Wang, Terry Van Dyke, Mark Vitucci, Andrea M. Werneke, Byron Huff, Ryan E. Bash, and Natalie O. Karpinich
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cell cycle checkpoint ,Blotting, Western ,Apoptosis ,Receptor tyrosine kinase ,Proto-Oncogene Proteins p21(ras) ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Movement ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Phosphoinositide 3-kinase ,biology ,Effector ,Brain Neoplasms ,fungi ,Cell Cycle ,PTEN Phosphohydrolase ,Cell cycle ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,Oncology ,Astrocytes ,Basic and Translational Investigations ,biology.protein ,Neurology (clinical) ,Signal transduction ,Mitogen-Activated Protein Kinases ,Glioblastoma ,Signal Transduction - Abstract
Glioblastoma (GBM) genomes feature recurrent genetic alterations that dysregulate core intracellular signaling pathways, including the G1/S cell cycle checkpoint and the MAPK and PI3K effector arms of receptor tyrosine kinase (RTK) signaling. Elucidation of the phenotypic consequences of activated RTK effectors is required for the design of effective therapeutic and diagnostic strategies.Genetically defined, G1/S checkpoint-defective cortical murine astrocytes with constitutively active Kras and/or Pten deletion mutations were used to systematically investigate the individual and combined roles of these 2 RTK signaling effectors in phenotypic hallmarks of glioblastoma pathogenesis, including growth, migration, and invasion in vitro. A novel syngeneic orthotopic allograft model system was used to examine in vivo tumorigenesis.Constitutively active Kras and/or Pten deletion mutations activated both MAPK and PI3K signaling. Their combination led to maximal growth, migration, and invasion of G1/S-defective astrocytes in vitro and produced progenitor-like transcriptomal profiles that mimic human proneural GBM. Activation of both RTK effector arms was required for in vivo tumorigenesis and produced highly invasive, proneural-like GBM.These results suggest that cortical astrocytes can be transformed into GBM and that combined dysregulation of MAPK and PI3K signaling revert G1/S-defective astrocytes to a primitive gene expression state. This genetically-defined, immunocompetent model of proneural GBM will be useful for preclinical development of MAPK/PI3K-targeted, subtype-specific therapies.
- Published
- 2013
17. Adrenomedullin gene dosage correlates with tumor and lymph node lymphangiogenesis
- Author
-
Kathleen M. Caron, Scott T. Espenschied, Yuri Fedoriw, Natalie O. Karpinich, Daniel O. Kechele, and Helen H. Willcockson
- Subjects
Vascular Endothelial Growth Factor A ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Gene Dosage ,Biology ,Biochemistry ,Metastasis ,Research Communications ,Adrenomedullin ,Carcinoma, Lewis Lung ,Mice ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Lymphangiogenesis ,Molecular Biology ,Lymph node ,Cell Proliferation ,Macrophages ,Lewis lung carcinoma ,medicine.disease ,Endothelial stem cell ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Lymphatic system ,Lymphatic Metastasis ,Female ,RNA Interference ,Lymph Nodes ,Biotechnology - Abstract
Adrenomedullin (AM) is a potent lymphangiogenic factor that promotes lymphatic endothelial cell (LEC) proliferation through a pharmacologically tractable G-protein-coupled receptor. Numerous types of human cancers have increased levels of AM; however, the functional consequences of this fact have not been characterized. Therefore, we evaluated whether modulating adrenomedullin (Adm) gene dosage within tumor cells affects lymphangiogenesis. Murine Lewis lung carcinoma (LLC) cells that overexpress or underexpress Adm were injected subcutaneously into C57BL/6 mice, and tumors were evaluated for growth and vascularization. A dosage range from ∼10 to 200% of wild-type Adm expression did not affect LLC proliferation in vitro or in vivo, nor did it affect angiogenesis. Notably, the dosage of Adm markedly and significantly influenced tumor lymphangiogenesis. Reduced Adm expression in tumors decreased the proliferation of LECs and the number of lymphatic vessels, while elevated tumor Adm expression led to enlarged lymphatic vessels. Moreover, overexpression of Adm in tumors induced sentinel lymph node lymphangiogenesis and led to an increased incidence of Ki67-positive foci within the lung. These data show that tumor-secreted AM is a critical factor for driving both tumor and lymph node lymphangiogenesis. Thus, pharmacological targeting of AM signaling may provide a new avenue for inhibition of tumor lymphangiogenesis.—Karpinich, N. O., Kechele, D. O., Espenschied, S. T., Willcockson, H. H., Fedoriw, Y., Caron, K. M. Adrenomedullin gene dosage correlates with tumor and lymph node lymphangiogenesis.
- Published
- 2012
18. Modeling Astrocytomas in a Family of Inducible Genetically Engineered Mice: Implications for Preclinical Cancer Drug Development
- Author
-
Elizabeth Bullitt, Qian Zhang, Terry Van Dyke, C. Ryan Miller, Serguei Kozlov, and Natalie O. Karpinich
- Subjects
endocrine system diseases ,business.industry ,Cell ,Astrocytoma ,Pharmacology ,medicine.disease ,medicine.disease_cause ,Phenotype ,nervous system diseases ,Pre-clinical development ,Clinical trial ,medicine.anatomical_structure ,nervous system ,Genetically Engineered Mouse ,medicine ,Cancer research ,Signal transduction ,business ,Carcinogenesis ,neoplasms - Abstract
Astrocytomas, the most common intracranial malignancies, are a morphologically and molecularly heterogeneous group of brain tumors with potentially dismal patient outcomes for which few effective drugs are available. Genetically engineered mouse (GEM) models of astrocytoma represent a powerful technique for defining the molecular and genetic abnormalities that contribute to tumorigenesis. Based on the genetic aberrations observed in human astrocytomas, we have generated a series of conditional, inducible GEM models of astrocytomas that recapitulate the spectrum of morphological phenotypes of human astrocytomas. However, the extent to which any given GEM model recapitulates the molecular alterations in human tumors must be determined to validate its usefulness in preclinical studies. We are currently pursuing comparative evaluation of primary astrocytomas as formed in GEM and in patients to (1) examine the signaling pathway abnormalities caused by defined genetic lesions in GEM astrocytomas and (2) identify protein biomarkers that can define human astrocytomas that most closely resemble their murine counterparts. To utilize these GEM for combined preclinical evaluation of targeted therapeutic agents and biomarkers predictive of response, we have developed a panel of cell-based assays (CBA) and an orthotopic allograft model of high-grade astrocytomas using primary astrocytes derived from GEM. These tools should prove useful for preclinical drug development studies and provide a link between preclinical drug development in GEM astrocytoma models and rational design of human clinical trials involving only those patients with tumors having similar signaling pathway abnormalities.
- Published
- 2009
- Full Text
- View/download PDF
19. Detailing the role of Bax translocation, cytochrome c release, and perinuclear clustering of the mitochondria in the killing of HeLa cells by TNF
- Author
-
Manuela Indelicato, Matteo Antonio Russo, Bruna Pucci, John L. Farber, Natalie O. Karpinich, Francesca Romana Bertani, and Marco Tafani
- Subjects
Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone ,Programmed cell death ,Oligomycin ,Physiology ,Cell Survival ,Clinical Biochemistry ,Apoptosis ,Mitochondrion ,Mitochondrial Membrane Transport Proteins ,Permeability ,chemistry.chemical_compound ,Adenosine Triphosphate ,Chloride Channels ,Furosemide ,Humans ,Cycloheximide ,Enzyme Inhibitors ,bcl-2-Associated X Protein ,biology ,ATP synthase ,Mitochondrial Permeability Transition Pore ,Tumor Necrosis Factor-alpha ,Uncoupling Agents ,Cytochrome c ,Cytochromes c ,Cell Biology ,Hydrogen-Ion Concentration ,Molecular biology ,Trifluoperazine ,Cell biology ,Mitochondria ,Protein Transport ,Cell killing ,chemistry ,Mitochondrial permeability transition pore ,Mitochondrial Membranes ,biology.protein ,Cyclosporine ,Oligomycins ,Bongkrekic Acid ,HeLa Cells - Abstract
Induction of cell death in HeLa cells with TNF and cycloheximide (CHX) required an adequate ATP supply and was accompanied by decrease in intracellular pH, translocation of Bax, perinuclear clustering of the mitochondria, and cytochrome c release. The chloride channel inhibitor furosemide prevented the intracellular acidification, the translocation of Bax and the cell death. Cyclosporin A (CyA) or bongkrekic acid (BK) inhibited the induction of the MPT, the release of cytochrome c and the cell death without affecting the perinuclear clustering of the mitochondria or the translocation of Bax. Energy depletion with the ATP synthase inhibitor oligomycin or the uncoupler FCCP in the presence of 2-deoxy-glucose prevented the perinuclear clustering of the mitochondria and the cell killing. However, mitochondrial translocation of Bax was still observed. By contrast, cytochrome c was released in the oligomycin-treated cells but not in the same cells treated with FCCP. The data demonstrate that apoptosis in HeLa cells is ATP dependent and requires the translocation of Bax. The movement of Bax to the mitochondria occurs before and during the perinuclear clustering of these organelles and does not require the presence of ATP. The release of cytochrome c depends on the induction of the mitochondrial permeability transition but not ATP content.
- Published
- 2008
20. The course of etoposide-induced apoptosis in Jurkat cells lacking p53 and Bax
- Author
-
Timothy G. Schneider, Matteo Antonio Russo, Natalie O. Karpinich, John L. Farber, and Marco Tafani
- Subjects
Programmed cell death ,Cytochrome ,Physiology ,T-Lymphocytes ,Clinical Biochemistry ,Apoptosis ,Jurkat cells ,Amino Acid Chloromethyl Ketones ,Jurkat Cells ,Cyclosporin a ,Humans ,Genes, Tumor Suppressor ,Phosphorylation ,Proto-Oncogene Proteins c-abl ,neoplasms ,Etoposide ,bcl-2-Associated X Protein ,biology ,Cytochrome c ,Tumor Suppressor Proteins ,Cytochromes c ,Nuclear Proteins ,Tumor Protein p73 ,Cell Biology ,DNA, Neoplasm ,Antineoplastic Agents, Phytogenic ,Caspase Inhibitors ,Cell biology ,Mitochondria ,DNA-Binding Proteins ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,Cell killing ,Mitochondrial permeability transition pore ,Proto-Oncogene Proteins c-bcl-2 ,Caspases ,Mitochondrial Membranes ,biology.protein ,Cyclosporine ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor Protein p53 ,Mitochondrial Swelling ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage - Abstract
Jurkat T-lymphocytes lack p53 and Bax but contain p73 and Bid and are killed by etoposide (ETO). With ETO c-abl is phosphorylated and phosphorylated p73 increased. Translocation of full-length Bid to mitochondria follows, with induction of the mitochondrial permeability transition (MPT) and release of cytochrome c into the cytosol. Pronounced swelling of mitochondria was evident ultrastructurally, and the MPT inhibitor cyclosporin A prevented the release of cytochrome c. Overexpression of Bcl-2 prevented the translocation of Bid, the release of cytochrome c, and cell death. The pan-caspase inhibitor ZVAD-FMK prevented the cell killing, but not the initial release of cytochrome c. An accumulation of tBid occurred at later times in association with Bid degradation. A sequence is proposed that couples DNA damage to Bid translocation via activation of c-abl and p73. Bid translocation induces the MPT, the event that causes release of cytochrome c, activation of caspases, and cell death.
- Published
- 2006
21. Re-evaluation of the distinction between type I and type II cells: The necessary role of the mitochondria in both the extrinsic and intrinsic signaling pathways upon fas receptor activation
- Author
-
Matteo Antonio Russo, Natalie O. Karpinich, John L. Farber, Ada Serroni, and Marco Tafani
- Subjects
Cell Survival ,Physiology ,Clinical Biochemistry ,Apoptosis ,Mitochondrion ,Biology ,Jurkat cells ,Cell Line ,Jurkat Cells ,Cytosol ,Cyclosporin a ,Humans ,fas Receptor ,Caspase 8 ,Cell Death ,Antibodies, Monoclonal ,Cytochromes c ,Cell Biology ,Fas receptor ,Caspase Inhibitors ,Mitochondria ,Cell biology ,Cell killing ,Cell culture ,Cyclosporine ,Bongkrekic Acid ,Oligopeptides ,Signal Transduction - Abstract
Cyclosporin A (CyA) and bongkrekic acid (BK) prevented Fas-induced apoptosis in two type I cell lines (H9 and SKW6.4) and two type II cell lines (Jurkat and CEM). CyA and BK inhibited the release of cytochrome c in all four cell lines. In type I cells and in CEM cells, CyA and BK did not prevent the translocation of Bax to the mitochondria. In these same cells, full-length Bid decreased in the mitochondria and cytosol. The cleavage product of Bid, tBid, appeared in the cytosol and to a lesser extent in the mitochondria. In Jurkat cells, Bid also decreased in the cytosol, but increased in the mitochondria. Similar to the other cells, tBid appeared in the mitochondria and cytosol. In the type I H9 and SKW6.4 cells and type II Jurkat cells, the caspase-8 inhibitor Z-Ile-Glu(OMe)-Thr-Asp(OMe)-CH2F (IETD) prevented the cell killing. In the type I cells, IETD prevented the translocation of Bax, the degradation of Bid and the accumulation of tBid. By contrast, IETD only marginally protected the type II CEM cells. In these cells in the presence of IETD, Bax translocated to the mitochondria, in the absence of any degradation of Bid or accumulation of tBid. In the type I H9 cells, IETD produced a depletion of ATP, an effect that did not occur in the type II CEM cells. It is concluded that in type I cells the extrinsic signaling pathway is mitochondrial dependent to the same extent as is the intrinsic pathway in type II cells.
- Published
- 2006
22. Cytochrome c release upon Fas receptor activation depends on translocation of full-length bid and the induction of the mitochondrial permeability transition
- Author
-
Marco Tafani, Timothy G. Schneider, Matteo Antonio Russo, Natalie O. Karpinich, John L. Farber, John G. Pastorino, and Kathryn A. Hurster
- Subjects
Time Factors ,Cell Survival ,Blotting, Western ,Cytochrome c Group ,Mitochondrion ,Biology ,Biochemistry ,Cell Line ,Membrane Potentials ,Jurkat Cells ,Malate Dehydrogenase ,Cyclosporin a ,Humans ,fas Receptor ,Molecular Biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cytochrome c ,Membrane Proteins ,Cell Biology ,Fas receptor ,Molecular biology ,Actins ,Mitochondria ,Microscopy, Electron ,Protein Transport ,Cell killing ,bcl-2 Homologous Antagonist-Killer Protein ,Mitochondrial permeability transition pore ,Apoptosis ,PARP inhibitor ,biology.protein ,Poly(ADP-ribose) Polymerases ,Carrier Proteins ,BH3 Interacting Domain Death Agonist Protein ,HeLa Cells ,Protein Binding - Abstract
In Jurkat cells Bid was cleaved upon activation of the Fas receptor with an anti-Fas antibody. The caspase-8 inhibitor benzyloxycarbonyl-Ile-Glu(OMe)-Thr-Asp(OMe)-CH(2)F (IETD) prevented the cleavage of Bid and the loss of viability. The nuclear enzyme poly(ADP-ribose)polymerase (PARP) was also cleaved upon the activation of caspases, and IETD similarly prevented PARP cleavage. The PARP inhibitor 3-aminobenzamide (3-AB) restored the cell killing in the presence of IETD, an effect that occurred without restoration of the cleavage of Bid or PARP. In the presence of 3-AB and IETD, translocation occurred of full-length Bid to the mitochondria. The induction of the mitochondrial permeability transition (MPT) was documented by the cyclosporin A (CyA) sensitivity of the release of cytochrome c, the release of malate dehydrogenase from the mitochondrial matrix, the loss of the mitochondrial membrane potential, and the pronounced swelling of these organelles, as assessed by electron microscopy. In addition to preventing all evidence of the MPT, CyA prevented the loss of cell viability, without effect on the cleavage of either Bid or PARP. The prevention of PARP cleavage by inhibition of caspase-3 resulted in a 10-fold activation of the enzyme and a resultant depletion of NAD and ATP. The PARP inhibitor 3-AB prevented the loss of NAD and ATP. Depletion of ATP by metabolic inhibitors similarly prevented the cell killing. It is concluded that the cleaving of PARP in Fas-mediated apoptosis allowed expression of an energy-dependent cell death program that included the translocation of full-length Bid to the mitochondria with induction of the MPT.
- Published
- 2002
23. Abstract 4305: Dissecting the requirements for astrocytoma and invasion using genetically-engineered mouse models
- Author
-
Ralf S. Schmid, Natalie O. Karpinich, Byron Huff, Mark Vitucci, Ryan E. Bash, and C. Ryan Miller
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,Kinase ,Astrocytoma ,Cancer ,Video microscopy ,medicine.disease ,medicine.disease_cause ,Oncology ,medicine ,biology.protein ,Cancer research ,Doubling time ,PTEN ,KRAS ,PI3K/AKT/mTOR pathway - Abstract
Astrocytomas are characterized by diffuse invasion, precluding their complete surgical resection. PTEN, a negative PI3 kinase (PI3K) pathway regulator, is altered in 40-80% of high-grade astrocytomas (HGA), including glioblastoma (GBM). However, its role in astrocytoma invasion remains unclear. Primary astrocytes from six genetically-engineered mouse (GEM) models, with conditional alleles that inactivate Rb (T) and/or Pten (P) and/or constitutively activate Kras (R, KrasG12D) upon Cre recombination, were used to analyze PI3K pathway signaling, proliferation, migration, and invasion in vitro by immunoblot, cell counting, wound healing and time-lapse video microscopy, and collagen invasion, respectively. Gene expression microarrays were used to compare the transcriptomes of GEM astrocytes to human HGA. Tumorigenicity and survival were determined in vivo in orthotopic allograft models. Invasion was assessed by morphometric analysis. Pten ablation increased levels of phospho-Akt and phospho-S6. In cells with both Rb inactivation and Kras activation (TR), complete inactivation of Pten shortened doubling time (DT) by 42% (P Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4305. doi:1538-7445.AM2012-4305
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.