Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res Treat ; 49(2): 374-386, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27488871

ABSTRACT

PURPOSE: When integrating molecularly targeted compounds in radiotherapy, synergistic effects of the systemic agent and radiation may extend the limits of patient tolerance, increasing the demand for understanding the pathophysiological mechanisms of treatment toxicity. In this Pelvic Radiation and Vorinostat (PRAVO) study, we investigated mechanisms of adverse effects in response to the histone deacetylase (HDAC) inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) when administered as a potential radiosensitiser. MATERIALS AND METHODS: This phase I study for advanced gastrointestinal carcinoma was conducted in sequential patient cohorts exposed to escalating doses of vorinostat combined with standard-fractionated palliative radiotherapy to pelvic target volumes. Gene expression microarray analysis of the study patient peripheral blood mononuclear cells (PBMC) was followed by functional validation in cultured cell lines and mice treated with SAHA. RESULTS: PBMC transcriptional responses to vorinostat, including induction of apoptosis, were confined to the patient cohort reporting dose-limiting intestinal toxicities. At relevant SAHA concentrations, apoptotic features (annexin V staining and caspase 3/7 activation, but not poly-(ADP-ribose)-polymerase cleavage) were observed in cultured intestinal epithelial cells. Moreover, SAHA-treated mice displayed significant weight loss. CONCLUSION: The PRAVO study design implemented a strategy to explore treatment toxicity caused by an HDAC inhibitor when combined with radiotherapy and enabled the identification of apoptosis as a potential mechanism responsible for the dose-limiting effects of vorinostat. To the best of our knowledge, this is the first report deciphering mechanisms of normal tissue adverse effects in response to an HDAC inhibitor within a combined-modality treatment regimen.


Subject(s)
Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Histone Deacetylase Inhibitors/adverse effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Biomarkers , Cell Line , Clinical Trials, Phase I as Topic , Combined Modality Therapy , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/therapy , Gene Expression Profiling , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/therapeutic use , Intestines/pathology , Male , Mice , Middle Aged , Pelvis/radiation effects , Radiotherapy/methods , Rats , Transcriptome , Vorinostat
2.
BMC Cancer ; 16: 531, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27461218

ABSTRACT

BACKGROUND: The use of targeted agents to impel dual inhibition of anti-apoptotic mechanisms and mTOR-mediated pro-survival signaling in colorectal carcinoma (CRC) cell lines with KRAS or BRAF mutation has been shown to induce apoptosis, a timely result given CRC entities harboring such mutations are in need of new therapies. Since CRC comprises heterogeneous tumors with predominant hypoxic components, we investigated effects of an inhibitor of anti-apoptotic Bcl-2 family proteins (ABT-737) in combination with an mTOR inhibitor (AZD8055)-collectively referred to as combo-Rx, in hypoxic CRC cell lines. METHODS: Cell viability measures, expression of proteins implicated in apoptosis and MAPK/PI3K-AKT/mTOR pathway signaling, and profiling of composite kinase activities were undertaken in a panel of 14 cell lines. RESULTS: In hypoxic conditions, combo-Rx suppressed viability of 13 of the cell lines, albeit ABT-737 did not significantly potentiate the inhibitory effect of single-agent AZD8055 in six of the models. Hypoxic KRAS/PIK3CA-mutant HCT-116 and HCT-15 cell lines (both with low endogenous expression of the anti-apoptotic Mcl-1 protein and showing augmented inhibition of viability following the addition of ABT-737 to AZD8055) responded to combo-Rx by induction of apoptosis but with the simultaneous strong Mcl-1 up-regulation and activation of MAPK/PI3K-conducted signaling. In contrast, in hypoxic KRAS-mutant LoVo (devoid of PIK3CA mutation), BRAF/PIK3CA-mutant RKO, and wild-type Colo320DM cell lines (all with high endogenous Mcl-1 expression and being resistant to the additional effect of ABT-737 to AZD8055), combo-Rx did not elicit apoptotic or pro-survival responses. CONCLUSIONS: The concurrent inhibition of anti-apoptotic proteins and mTOR-mediated signaling in hypoxic KRAS/PIK3CA-mutant CRC cell lines resulted in pro-survival responses in parallel with the intended anti-proliferative effects, a finding that should be of note if considering combinatory targeting of multiple pathways in this CRC entity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Colorectal Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Hypoxia , Up-Regulation
3.
PLoS One ; 8(8): e72692, 2013.
Article in English | MEDLINE | ID: mdl-24023633

ABSTRACT

BACKGROUND: Treatment of metastatic malignant melanoma patients harboring BRAF(V600E) has improved drastically after the discovery of the BRAF inhibitor, vemurafenib. However, drug resistance is a recurring problem, and prognoses are still very bad for patients harboring BRAF wild-type. Better markers for targeted therapy are therefore urgently needed. METHODOLOGY: In this study, we assessed the individual kinase activity profiles in 26 tumor samples obtained from patients with metastatic malignant melanoma using peptide arrays with 144 kinase substrates. In addition, we studied the overall ex-vivo inhibitory effects of vemurafenib and sunitinib on kinase activity status. RESULTS: Overall kinase activity was significantly higher in lysates from melanoma tumors compared to normal skin tissue. Furthermore, ex-vivo incubation with both vemurafenib and sunitinib caused significant decrease in phosphorylation of kinase substrates, i.e kinase activity. While basal phosphorylation profiles were similar in BRAF wild-type and BRAF(V600E) tumors, analysis with ex-vivo vemurafenib treatment identified a subset of 40 kinase substrates showing stronger inhibition in BRAF(V600E) tumor lysates, distinguishing the BRAF wild-type and BRAF(V600E) tumors. Interestingly, a few BRAF wild-type tumors showed inhibition profiles similar to BRAF(V600E) tumors. The kinase inhibitory effect of vemurafenib was subsequently analyzed in cell lines harboring different BRAF mutational status with various vemurafenib sensitivity in-vitro. CONCLUSIONS: Our findings suggest that multiplex kinase substrate array analysis give valuable information about overall tumor kinase activity. Furthermore, intra-assay exposure to kinase inhibiting drugs may provide a useful tool to study mechanisms of resistance, as well as to identify predictive markers.


Subject(s)
Indoles/pharmacology , Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/enzymology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Aged , Cell Line, Tumor , Cluster Analysis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Male , Melanoma/pathology , Middle Aged , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Mutation/genetics , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/drug effects , Skin/enzymology , Skin/pathology , Skin Neoplasms/pathology , Substrate Specificity/drug effects , Sunitinib , Vemurafenib
4.
PLoS One ; 6(9): e24012, 2011.
Article in English | MEDLINE | ID: mdl-21915275

ABSTRACT

In cancer, combinations of drugs targeting different cellular functions is well accepted to improve tumor control. We studied the effects of a Pseudomonas exotoxin A (PE)-based immunotoxin, the 9.2.27PE, and the BH-3 mimetic compound ABT-737 in a panel of melanoma cell lines. The drug combination resulted in synergistic cytotoxicity, and the cell death observed was associated with apoptosis, as activation of caspase-3, inactivation of Poly (ADP-ribose) polymerase (PARP) and increased DNA fragmentation could be prevented by pre-treatment with caspase and cathepsin inhibitors. We further show that ABT-737 caused endoplasmic reticulum (ER) stress with increased GRP78 and phosphorylated eIF2α protein levels. Moreover, treatment with ABT-737 increased the intracellular calcium levels, an effect which was enhanced by 9.2.27PE, which as a single entity drug had minimal effect on calcium release from the ER. In addition, silencing of Mcl-1 by short hairpin RNA (shRNA) enhanced the intracellular calcium levels and cytotoxicity caused by ABT-737. Notably, the combination of 9.2.27PE and ABT-737 caused growth delay in a human melanoma xenograft mice model, supporting further investigations of this particular drug combination.


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Immunotoxins/therapeutic use , Melanoma/drug therapy , Nitrophenols/therapeutic use , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Biphenyl Compounds/pharmacology , Blotting, Western , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Fragmentation/drug effects , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Immunotoxins/pharmacology , Melanoma/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Nitrophenols/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
5.
J Immunother ; 34(5): 438-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21577141

ABSTRACT

The combined use of several drugs targeting different cellular functions is one approach to achieve tumor control in cancer. We studied the effects of Pseudomonas exotoxin A (PE)-based immunotoxins (ITs), the 9.2.27PE and the 425.3PE, together with 2 different triphenylmethyl derivatives, triphenylmethyl phosphonates and phosphonochloridates (TPMP)-I-2 and 4BI. Combining the triphenylmethyl derivatives with ITs enhanced the cytotoxic effect of the ITs, with TPMP-I-2 in combination with the 425.3PE (targeting the epidermal growth factor receptor) being the most promising combination. The cytotoxicity involving signs of apoptosis was observed in cancer cells from different origins in vitro. It is interesting to note that treatment with IT, TPMP-I-2, or 4BI alone or in combination resulted in strongly decreased protein levels of stearoyl-CoA desaturase. Stearoyl-CoA desaturase is the rate-limiting enzyme for converting saturated fatty acids into monounsaturated fatty acids needed for membrane genesis. Furthermore, the combination of 425.3PE and TPMP-I-2 prolonged the survival time of nude rats in a simulated micrometastatic cervical cancer model. In addition, we demonstrate that a combination of the 425.3PE and 4BI was more effective in reducing tumor growth in a breast cancer model in nude mice compared with either agent alone.


Subject(s)
ADP Ribose Transferases/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bacterial Toxins/pharmacology , Breast Neoplasms/drug therapy , Exotoxins/pharmacology , Immunotoxins/pharmacology , Skin Neoplasms/drug therapy , Stearoyl-CoA Desaturase/biosynthesis , Uterine Cervical Neoplasms/drug therapy , Virulence Factors/pharmacology , ADP Ribose Transferases/immunology , Animals , Apoptosis/drug effects , Bacterial Toxins/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Survival/drug effects , Drug Synergism , ErbB Receptors/metabolism , Exotoxins/immunology , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Organophosphonates/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Rats , Rats, Nude , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , Stearoyl-CoA Desaturase/genetics , Trityl Compounds/pharmacology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/mortality , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
6.
J Immunother ; 33(3): 272-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20445347

ABSTRACT

We have earlier shown that the 9.2.27 Pseudomonas Exotoxin A (PE) immunotoxin (IT) efficiently kills melanoma cells through inhibition of protein synthesis followed by some morphologic and biochemical features of apoptosis, a different cell killing mechanism than the one caused by Dacarbazine (DTIC), a chemotherapeutic drug used to treat malignant melanoma. To examine whether induced DTIC resistance also is a determining factor for the effectiveness of 9.2.27PE IT, we developed a DTIC resistant subline, FEMX-200DR, from the DTIC sensitive cell line FEMX. The cell variants were treated with 9.2.27PE, an IT binding to the high molecular weight-melanoma associated antigen (HMW-MAA) expressed on most malignant melanoma cells. The IT was equally effective in killing the FEMX-200DR and the FEMX cells, and the cell death was primarily caused by inhibition of protein synthesis. The DNA repair enzyme and apoptotic marker PARP, a substrate of caspase-3, was inactivated, although we observed only a minor activation of caspase-3 and caspase-8, intracellular proteases involved in apoptosis. In addition to being DTIC resistant, the FEMX-200DR cells were also more resistant to apoptosis than the parent cells as a 3 times higher concentration of the apoptotic inducer Staurosporine was needed to obtain IC50. Furthermore, in early passage malignant melanoma cell lines established from lymph node metastases, the 9.2.27PE caused a time-dependent and dose-dependent decrease in cell viability independent of their DTIC sensitivity. These findings show that the 9.2.27PE IT efficiently can cause cell death in malignant melanoma cells independent of their level of resistance to apoptosis and DTIC.


Subject(s)
Dacarbazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Immunotoxins/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/immunology , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Oligonucleotide Array Sequence Analysis , Poly(ADP-ribose) Polymerases/metabolism , Protein Biosynthesis/drug effects , Reverse Transcriptase Polymerase Chain Reaction
7.
Ochsner J ; 10(3): 193-9, 2010.
Article in English | MEDLINE | ID: mdl-21603377

ABSTRACT

The incidence of melanoma is rising in the Western population, and melanoma is the most aggressive form of skin cancer with a very poor prognosis once it has progressed to metastatic stages. Patients with stage IV melanoma (metastases to distant lymph nodes and other areas of the body) are treated with the chemotherapeutic drug dacarbazine (DTIC). However, fewer than 5% of the patients treated with DTIC sustain long-term complete responses; hence, DTIC is administered with palliative purposes. New therapy is urgently needed. We are developing another therapeutic strategy, specifically targeting melanoma cells with the 9.2.27PE immunotoxin (IT). ITs bind to antigens overexpressed on cancer cells and are therefore tumor selective. This targeted approach may potentially cause fewer side effects in a clinical situation compared to conventional approaches like chemotherapy and radiotherapy.

8.
Int J Cancer ; 125(1): 23-33, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19350633

ABSTRACT

Malignant melanomas are generally drug resistant and have a very poor prognosis. We have studied the effects of a chemical conjugate of pseudomonas exotoxin A (PE) and the antibody 9.2.27, which recognizes the high molecular weight melanoma associated antigen (HMW-MAA) expressed in most malignant melanomas and melanoma cell lines. We demonstrate that the 9.2.27PE immunotoxin (IT) induces cell death in malignant melanoma cells through protein synthesis inhibition followed by some morphological and biochemical features of apoptosis, like rounding up of cells, chromatin condensation and inactivation of PARP. Unlike previous results with the 425.3PE IT in breast cancer cells, we detected no depolarization of the mitochondrial membrane after 9.2.27PE IT treatment. This is likely due to the lack of strong activation of caspase-8 and caspase-3. The lack of depolarization suggests that cytochrome c, a molecule that triggers activation of caspase-3, was retained within the mitochondria. In addition, the protein level of the antiapoptotic Bcl-2 did not decrease in contrast to other antiapoptotic molecules belonging to the inhibitor of apoptosis and the Bcl-2 family. This suggests that Bcl-2 may play a role in maintaining the mitochondrial membrane integrity in the 9.2.27PE-treated cells. Nevertheless, 9.2.27PE IT efficiently killed malignant melanoma cells that can be ascribed to inhibition of protein synthesis followed by some morphological and biochemical features of apoptosis.


Subject(s)
ADP Ribose Transferases/pharmacology , Antigens, Neoplasm/immunology , Apoptosis/drug effects , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Immunotoxins/pharmacology , Melanoma/pathology , Virulence Factors/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Chromatin/metabolism , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Melanoma/enzymology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...