Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Endocrinol Invest ; 44(9): 1927-1933, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33576954

ABSTRACT

BACKGROUND AND PURPOSE: Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that enhance the immune response against cancer cells. ICIs are generally well tolerated, although endocrine immune-related adverse events (irAEs) are common. We investigated the risk factors for thyroid irAEs in patients treated with ICIs. Moreover, we evaluated the clinical outcome of subjects who became hypothyroid compared to euthyroid patients. PATIENTS AND METHODS: We retrospectively analyzed a series of 195 consecutively subjects treated with ICIs for metastatic tumors at the University of Naples "Federico II" between January 2014 and March 2020. Only subjects tested for thyroid function before and during the treatment with ICIs were included. RESULTS: In the 96 patients treated with ICIs who were included [66 males, median age: 62 years (27-87)], thyroid irAEs occurred in 36 (37.5%), 16 (16.7%) a transient thyrotoxicosis, and 20 (20.8%) an hypothyroidism (in nine subjects hypothyroidism was preceded by a transient thyrotoxicosis). Only baseline TSH levels above 1.67 mIU/L and positive anti-thyroid antibodies (Ab-T) were associated with a higher risk of hypothyroidism. Patients with hypothyroidism during ICI treatment showed an improved 2-year PFS (HR = 0.82 CI 0.47-1.43; p = 0.0132) and OS (HR = 0.38 CI 95% 0.17-0.80; p = 0.011) compared to euthyroid patients. CONCLUSIONS: Baseline TSH levels above 1.67 mIU/L and presence of Ab-T are risk factors for the development of thyroid irAEs. Patients affected by thyroid irAEs showed a longer survival than patients who remained euthyroid.


Subject(s)
Hypothyroidism/blood , Hypothyroidism/etiology , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Neoplasms/complications , Thyrotropin/blood , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/adverse effects , Female , Humans , Hypothyroidism/epidemiology , Immune Checkpoint Inhibitors/therapeutic use , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasms/mortality , Neoplasms/therapy , Progression-Free Survival , Retrospective Studies , Risk Factors , Survival Analysis , Thyroid Function Tests , Thyrotoxicosis/epidemiology , Treatment Outcome
2.
Clin Oncol (R Coll Radiol) ; 29(11): e186-e194, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28803687

ABSTRACT

AIMS: To identify predictors of asymptomatic radiation-induced abdominal atherosclerosis in patients treated with radiotherapy and evaluated by abdominal vascular ultrasonography. MATERIALS AND METHODS: Forty-two testicular classic seminoma patients (median age 34 years, range 16-56) undergoing radical inguinal orchiectomy were analysed. Twenty-six patients underwent post-surgery radiotherapy (median total dose 25 Gy, range 25-43), two of them also received chemotherapy (CHT) and 16 patients were treated with surgery alone or by surgery followed by CHT (control group). The presence of stenosis in an abdominal vessel and renal resistive index (RRI), evaluated by echo-colour Doppler (ECD), were considered as indicators of late vascular damage. Chi-square and Mann-Whitney tests were used to compare groups. For the radiotherapy group, near maximum (D2%) and mean dose (Dmean) metrics of critical structures (abdominal arteries and renal hila) were extracted from retrievable dose maps (18 of 26 radiotherapy patients). To evaluate clinical and dosimetric factors associated with vascular damage, univariate and multivariate analyses were carried out. The impact of dose to arteries, evaluated as separate subvolumes, was analysed comparing the stenotic arteries with normal ones by logistic regression. The area under the receiver operator characteristic curve (AUC) was used to evaluate the test accuracy. RESULTS: In the radiotherapy group there was a significantly different incidence of stenosis (31% versus 0%, P = 0.016) and a higher median average RRI (0.63 versus 0.60, P = 0.032) compared with the control group. The median time intervals between treatment and ECD were 64 months (range 12-120) and 48 months (range 12-168) in the radiotherapy and control groups (P = 0.399), respectively. A younger age at radiotherapy was the only clinical risk factor for stenosis (P = 0.006). Artery Dmean was significantly associated with stenosis (P = 0.008), with an odds ratio of 1.13 (95% confidence interval 1.01-1.26) and an AUC of 0.85 (95% confidence interval 0.77-0.91). Renal hilum D2% was correlated with RRI (Rs = 0.406, P = 0.02). CONCLUSIONS: Late vascular damage represents a potential effect of abdominal radiotherapy, even at a moderate dose. Younger age at irradiation, artery and renal hila dose metrics are associated with increased risk. Ultrasound-based follow-up may allow for non-invasive early detection of asymptomatic radiation-induced damage, helping to prevent severe vascular events.


Subject(s)
Abdomen/radiation effects , Atherosclerosis/chemically induced , Abdomen/diagnostic imaging , Adolescent , Adult , Atherosclerosis/pathology , Female , Humans , Incidence , Male , Middle Aged , Ultrasonography , Young Adult
4.
Br J Cancer ; 109(3): 686-93, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23839492

ABSTRACT

BACKGROUND: We aimed to study key signalling proteins involved in angiogenesis and proliferation on the response to inhibitors of tyrosine kinases and mammalian target of rapamycin in first- and in second-line treatment of renal cell carcinoma (RCC). METHODS: In a panel of human RCC tumours, in vitro and in nude mice, we evaluated the effect of sunitinib, sorafenib and everolimus, alone and in sequence, on tumour growth and expression of signalling proteins involved in proliferation and resistance to treatment. RESULTS: We demonstrated that, as single agents, sunitinib, sorafenib and everolimus share similar activity in inhibiting cell proliferation, signal transduction and vascular endothelial growth factor (VEGF) secretion in different RCC models, both in vitro and in tumour xenografts. Pre-treatment with sunitinib reduced the response to subsequent sunitinib and sorafenib but not to everolimus. Inability by sunitinib to persistently inhibit HIF-1, VEGF and pMAPK anticipated treatment resistance in xenografted tumours. After first-line sunitinib, second-line treatment with everolimus was more effective than either sorafenib or rechallenge with sunitinib in interfering with signalling proteins, VEGF and interleukin-8, translating into a significant advantage in tumour growth inhibition and mice survival. CONCLUSION: We demonstrated that a panel of angiogenic and signalling proteins can correlate with the onset of resistance to sunitinib and the activity of everolimus in second line.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiogenic Proteins/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenic Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Growth Processes/drug effects , Everolimus , Humans , Indoles/administration & dosage , Indoles/pharmacology , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/administration & dosage , Pyrroles/pharmacology , Random Allocation , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sorafenib , Sunitinib , Xenograft Model Antitumor Assays
5.
Br J Cancer ; 108(8): 1616-23, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23571736

ABSTRACT

BACKGROUND: Targeting the mammalian target of rapamycin by everolimus is a successful approach for renal cell carcinoma (RCC) therapy. The Toll-like receptor 9 agonist immune modulatory oligonucleotide (IMO) exhibits direct antitumour and antiangiogenic activity and cooperates with both epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) inhibitors. METHODS: We tested the combination of IMO and everolimus on models of human RCC with different Von-Hippel Lindau (VHL) gene status, both in vitro and in nude mice. We studied their direct antiangiogenic effects on human umbilical vein endothelial cells. RESULTS: Both IMO and everolimus inhibited in vitro growth and survival of RCC cell lines, and their combination produced a synergistic inhibitory effect. Moreover, everolimus plus IMO interfered with EGFR-dependent signaling and reduced VEGF secretion in both VHL wild-type and mutant cells. In RCC tumour xenografts, IMO plus everolimus caused a potent and long-lasting cooperative antitumour activity, with reduction of tumour growth, prolongation of mice survival and inhibition of signal transduction. Furthermore, IMO and everolimus impaired the main endothelial cell functions. CONCLUSION: A combined treatment with everolimus and IMO is effective in VHL wild-type and mutant models of RCC by interfering with tumour growth and angiogenesis, thus representing a potentially effective, rationale-based combination to be translated in the clinical setting.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Oligonucleotides/pharmacology , Sirolimus/analogs & derivatives , Toll-Like Receptor 9/agonists , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Synergism , Everolimus , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Oligonucleotides/genetics , Oligonucleotides/immunology , Random Allocation , Sirolimus/pharmacology , Toll-Like Receptor 9/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xenograft Model Antitumor Assays
6.
Br J Cancer ; 102(3): 513-9, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20051960

ABSTRACT

BACKGROUND: Overexpression of ErbB2 receptor in breast cancer is associated with disease progression and poor prognosis. Trastuzumab, the only humanised anti-ErbB2 antibody currently used in breast cancer, has proven to be effective; however, a relevant problem for clinical practice is that a high fraction of breast cancer patients shows primary or acquired resistance to trastuzumab treatment. METHODS: We tested on trastuzumab-resistant cells two novel human anti-tumour immunoconjugates engineered in our laboratory by fusion of a human anti-ErbB2 scFv, termed Erbicin, with either a human RNase or the Fc region of a human IgG1. Both Erbicin-derived immunoagents (EDIAs) are selectively cytotoxic for ErbB2-positive cancer cells in vitro and vivo, target an ErbB2 epitope different from that recognised by trastuzumab and do not show cardiotoxic effects. RESULTS: We report that EDIAs are active also on trastuzumab-resistant tumour cells both in vitro and in vivo, most likely because of the different epitope recognised, as EDIAs, unlike trastuzumab, were found to be able to inhibit the signalling pathway downstream of ErbB2. CONCLUSION: These results suggest that EDIAs are immunoagents that could not only fulfil the therapeutic need of patients ineligible to trastuzumab treatment due to cardiac dysfunction but also prove to be useful for breast cancer patients unresponsive to trastuzumab treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Drug Resistance, Neoplasm , Epitopes , Female , Humans , Immunoglobulin G/therapeutic use , Mice , Mice, Inbred BALB C , Ribonucleases/therapeutic use , Trastuzumab , Xenograft Model Antitumor Assays
7.
Int J Immunopathol Pharmacol ; 23(4): 1275-9, 2010.
Article in English | MEDLINE | ID: mdl-21244779

ABSTRACT

Thymomas are rare tumors, which can be associated to a variety of paraneoplastic syndromes, including a fatal hypogammaglobulinemia, namely Good?s Syndrome (GS). Although the combination of thymoma and hypogammaglobulinemia is regarded as sufficient for diagnosis of Good?s syndrome, some thymoma patients with a clear clinical picture of immunodeficiency present normal levels of immunoglobulins. We describe the case of a patient, with a 20-year history of thymoma, who underwent several operations and lines of chemotherapy, and suffered from recurrent infections, including one rare skin infection from Pseudoallescheria boydii. The patient constantly presented normal levels of gammaglobulins.


Subject(s)
Agammaglobulinemia/etiology , Paraneoplastic Syndromes/etiology , Thymoma/complications , Thymus Neoplasms/complications , Adult , Humans , Male , Thymoma/immunology , Thymus Neoplasms/immunology
8.
Int J Immunopathol Pharmacol ; 23(4): 1281-5, 2010.
Article in English | MEDLINE | ID: mdl-21244780

ABSTRACT

Paraneoplastic sensitive neuropathy is one of the most common presentations among a group of cancer-related disorders known as Paraneoplastic Neurological Syndromes (PNS). PNS likely have an autoimmune etiology since they have been associated with the presence of antibodies against neuronal antigens expressed by tumor cells (such as anti-Hu, anti-Ri and anti-Yo). The tumors most frequently associated with PSN and onconeural antibodies are lung cancer, lymphomas and gynaecological tumors; however, they have also been described in other tumors. We report, for the first time, a case of neuroendocrine tumor of duodenum and PNS associated with anti-Hu antibodies. Moreover, we analyze and discuss the clinical implications that PNS and anti-Hu could have in patients with tumors.


Subject(s)
Antibodies, Antinuclear/analysis , Duodenal Neoplasms/complications , ELAV Proteins/immunology , Neuroendocrine Tumors/complications , Paraneoplastic Polyneuropathy/etiology , Duodenal Neoplasms/immunology , Humans , Male , Middle Aged , Neuroendocrine Tumors/immunology
9.
Br J Cancer ; 99(3): 473-80, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18665191

ABSTRACT

We investigated the antitumour effect and ability to overcome the resistance to anti-EGFR drugs of enzastaurin, an inhibitor of VEGFR-dependent PKCbeta signalling. Enzastaurin was evaluated alone and in combination with the EGFR inhibitor gefitinib, on growth and signalling protein expression in human cancer cells sensitive and resistant to anti-EGFR drugs, both in vitro and in nude mice. We demonstrated the marked inhibitory activity of enzastaurin against GEO colon and PC3 prostate cancer cells and their gefitinib-resistant counterparts GEO-GR and PC3-GR, accompanied by inhibition of pAkt and its effector pp70S6K, pGSK3beta and VEGF expression and secretion. Moreover, enzastaurin showed a cooperative effect with gefitinib in parental and in gefitinib-resistant cells. Remarkably, these results were confirmed in vivo, where enzastaurin showed antitumour activity and cooperativity with gefitinib in mice grafted with GEO and GEO-GR tumours, incrementing their median survival and inhibiting the aforesaid protein expression and secretion in tumour specimens. In conclusion, enzastaurin by interfering with signalling proteins implicated in EGFR drug resistance markedly cooperates with gefitinib in sensitive and gefitinib-resistant tumours, thus overcoming and reverting such resistance and providing a rational basis for its development in patients resistant to anti-EGFR drugs.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Indoles/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Culture Media, Conditioned , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Gefitinib , Humans , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Quinazolines/pharmacology , Vascular Endothelial Growth Factor A/metabolism
10.
Br J Cancer ; 98(5): 923-30, 2008 Mar 11.
Article in English | MEDLINE | ID: mdl-18319715

ABSTRACT

Inhibition of a single transduction pathway is often inefficient due to activation of alternative signalling. The mammalian target of rapamycin (mTOR) is a key intracellular kinase integrating proliferation, survival and angiogenic pathways and has been implicated in the resistance to EGFR inhibitors. Thus, mTOR blockade is pursued to interfere at multiple levels with tumour growth. We used everolimus (RAD001) to inhibit mTOR, alone or in combination with anti-EGFR drugs gefitinib or cetuximab, on human cancer cell lines sensitive and resistant to EGFR inhibitors, both in vitro and in vivo. We demonstrated that everolimus is active against EGFR-resistant cancer cell lines and partially restores the ability of EGFR inhibitors to inhibit growth and survival. Everolimus reduces the expression of EGFR-related signalling effectors and VEGF production, inhibiting proliferation and capillary tube formation of endothelial cells, both alone and in combination with gefitinib. Finally, combination of everolimus and gefitinib inhibits growth of GEO and GEO-GR (gefitinib resistant) colon cancer xenografts, activation of signalling proteins and VEGF secretion. Targeting mTOR pathway with everolimus overcomes resistance to EGFR inhibitors and produces a cooperative effect with EGFR inhibitors, providing a valid therapeutic strategy to be tested in a clinical setting.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinases/drug effects , Sirolimus/analogs & derivatives , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Drug Resistance, Neoplasm , Endothelial Cells/drug effects , Everolimus , Gefitinib , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms/pathology , Neovascularization, Physiologic/drug effects , Quinazolines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transplantation, Heterologous
12.
Br J Cancer ; 92(9): 1644-9, 2005 May 09.
Article in English | MEDLINE | ID: mdl-15856038

ABSTRACT

The aim of the study was to assess the toxicity and the clinical activity of biweekly oxaliplatin in combination with infusional 5-fluorouracil (5-FU) and folinic acid (FA) administered every 2 weeks (FOLFOX-4 regimen) in patients with advanced gastric cancer (AGC). A total of 61 previously untreated AGC patients were treated with oxaliplatin 85 mg m(-2) on day 1, FA 200 mg m(-2) as a 2 h infusion followed by bolus 5-FU 400 mg m(-2) and a 22 h infusion of 5-FU 600 mg m(-2), repeated for 2 consecutive days every 2 weeks. All patients were assessable for toxicity and response to treatment. Four (7%) complete responses and 19 partial responses were observed (overall response rate, 38%). Stable disease was observed in 22 (36%) patients, with progressive disease in the other six (10%) patients. Median time to progression (TTP) and median overall survival (OS) were 7.1 and 11.2 months, respectively. National Cancer Institute Common Toxicity Criteria grade 3 and 4 haematologic toxicities were neutropenia, anaemia and thrombocytopenia in 36, 10 and 5% of the patients, respectively. Grade 3 peripheral neuropathy was recorded in three (5%) patients. FOLFOX-4 is an active and well-tolerated chemotherapy. Response rate (RR), TTP and OS were comparable with those of other oxaliplatin-based regimens, suggesting a role for this combination in gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/therapeutic use , Leucovorin/therapeutic use , Organoplatinum Compounds/therapeutic use , Stomach Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Signet Ring Cell/drug therapy , Drug Administration Schedule , Female , Fluorouracil/adverse effects , Humans , Leucovorin/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , Organoplatinum Compounds/adverse effects , Stomach Neoplasms/pathology , Survival Rate
13.
Endocr Relat Cancer ; 11(1): 51-68, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15027885

ABSTRACT

The currently available treatment of cancer patients is based on the use of cytotoxic drugs and/or of ionizing radiations, which have potent antitumor activity, but also cause clinically relevant side effects, since they affect cellular targets that are common to both cancer cells and normal proliferating cells. In the past 20 years, the discoveries on the molecular mechanisms of cancer development and progression have prompted the search for agents which are more selective for cancer cell molecular targets. The possibility of combining conventional cytotoxic drugs with novel agents that specifically interfere with key pathways controlling cancer cell survival, proliferation, invasion and/or metastatic spreading has generated a wide interest. This could be a promising therapeutic approach for several reasons. First, since the cellular targets for these agents and their mechanism(s) of action are different from those of cytotoxic drugs, it is possible for their combination with chemotherapy without cross-resistance. Second, alterations in the expression and/or the activity of genes that regulate mitogenic signals not only can directly cause perturbation of cell growth, but also may affect the sensitivity of cancer cells to conventional chemotherapy and radiotherapy. In this review, we will discuss the biologic bases of the combination of molecular targeted drugs with conventional medical cancer treatments and the available results of the first series of clinical trials in cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Signal Transduction/drug effects , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Combined Modality Therapy , ErbB Receptors/antagonists & inhibitors , Farnesyltranstransferase , Genes, bcl-2/drug effects , Humans , Neoplasms/therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Protein Kinase C/antagonists & inhibitors , Thionucleotides/pharmacology , ras Proteins/antagonists & inhibitors
14.
Ann Oncol ; 13(3): 392-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11996469

ABSTRACT

BACKGROUND: DNA damage caused by platinum agents is frequently followed by induction of topoisomerase I, providing a rationale for use of platinum-based compounds with topoisomerase I inhibitors. MATERIALS AND METHODS: We studied the effect of a sequential schedule of oxaliplatin on day I and topotecan on days 2-5, in human colon and ovarian cancer cells in vitro, in nude mice bearing human cancer xenografts and finally in cancer patients in a phase I trial. RESULTS: We demonstrated a supra-additive effect of this combination on inhibition of colony formation and induction of apoptosis in vitro. We then demonstrated that the two agents in combination markedly inhibit tumor growth in nude mice. We translated these results into a clinical setting, conducting a phase I study in cancer patients with oxaliplatin 85 mg/m2 on day 1 and topotecan at doses escalating from 0.5 to 1.5 mg/m2 on days 2-5. Sixty cycles of treatment were administered to 18 patients affected prevalently by ovarian and colorectal cancer. Combination with topotecan 1.5 mg/m2 caused a dose-limiting toxicity. Therefore the maximum tolerated dose of topotecan was 1.25 mg/m2, at which six patients experienced a mild hematological and gastrointestinal toxicity. We also obtained evidence of clinical activity, particularly in ovarian cancer. CONCLUSIONS: Our results provide a solid biological and clinical rationale for a phase II trial at the recommended doses of oxaliplatin 85 mg/m2 and topotecan 1.25 mg/m2, possibly in ovarian cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Tumor Cells, Cultured/drug effects , Adult , Aged , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Male , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Topotecan/administration & dosage
15.
Clin Cancer Res ; 7(12): 4156-63, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751516

ABSTRACT

PURPOSE: Protein kinase A type I (PKAI) and the epidermal growth factor receptor (EGFR) play a role in neoplastic transformation and interact with each other in transducing mitogenic signals. We developed different PKAI and EGFR inhibitors, demonstrating their cooperation with cytotoxic drugs and the therapeutic potential of the combined blockade of PKAI and EGFR. In this study, we investigated the effect of orally active PKAI and EGFR inhibitors in combination with a novel taxane. EXPERIMENTAL DESIGN: We combined a hybrid PKAI antisense oligonucleotide sequence (AS-PKAI), the EGFR inhibitor ZD1839 (Iressa), and the taxane IDN5109, studying their effect on human cancer growth, apoptosis, and angiogenesis and measuring vascular endothelial growth factor (VEGF) expression and vessel formation in vitro and after oral administration in nude mice. RESULTS: We demonstrated cooperative growth inhibitory and proapoptotic effects and inhibition of VEGF expression with any combination of two drugs and a marked synergistic effect when all three agents were combined. Oral administration of AS-PKAI, ZD1839, and IDN5109 in combination to nude mice caused a remarkable antitumor effect with no histological evidence of tumors in 50% of mice 5 weeks after treatment withdrawal, accompanied by complete suppression of vessel formation and VEGF expression. CONCLUSION: This is the first demonstration of the cooperative antitumor and antiangiogenic activity of three novel agents that block multiple signaling pathways after oral administration. Because all agents are under clinical evaluation in cancer patients, our results provide a rationale to translate this feasible therapeutic strategy in a clinical setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Cyclic AMP-Dependent Protein Kinases/genetics , Oligodeoxyribonucleotides, Antisense/therapeutic use , Ovarian Neoplasms/drug therapy , Quinazolines/therapeutic use , Administration, Oral , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Endothelial Growth Factors/genetics , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Lymphokines/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Oligodeoxyribonucleotides, Antisense/administration & dosage , Ovarian Neoplasms/pathology , Quinazolines/administration & dosage , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Clin Cancer Res ; 7(8): 2537-44, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489837

ABSTRACT

Protein kinase A type I (PKAI) plays a key role in neoplastic transformation, conveys mitogenic signals from different sources, and is overexpressed in the majority of human tumors. Inhibition of PKAI by different tools results in cancer-cell growth inhibition in vitro and in vivo. We and others have recently shown that a novel class of mixed-backbone oligonucleotides targeting the PKAI subunit RIalpha exhibits improved pharmacokinetic properties and antitumor activity accompanied by increased apoptosis in several human cancer types in vitro and in vivo. The role of bcl-2 in the control of apoptosis has been widely documented, and the inhibition of bcl-2 expression and function may have important therapeutic implications. In fact, oligonucleotides antisense bcl-2 have shown antitumor activity in animal models and have successfully completed early clinical trials. Recent studies have demonstrated a direct role of PKA in the regulation of the bcl-2-dependent apoptotic pathway. Therefore, we have investigated the combined blockade of PKA and bcl-2 by antisense strategy as a potential therapeutic approach. The novel hybrid DNA/RNA mixed-backbone oligonucleotide antisense RIalpha (AS RIalpha) in combination with the antisense bcl-2 (AS bcl-2), cooperatively inhibited bcl-2 expression and soft agar growth and induced apoptosis in different human cancer cell lines. p.o. administration of AS RIalpha in combination with i.p. AS bcl-2 caused a marked antitumor effect and a significant prolongation of survival in nude mice bearing human colon cancer xenografts. Moreover, histochemical analysis of tumor specimens showed inhibition of RIalpha and Ki67 expression, inhibition of angiogenesis, and parallel induction of apoptosis in vivo. The results of our study imply an interaction between the PKA and bcl-2 signaling pathways and, because both antisenses have now entered Phase II trials, provide the rationale to translate this novel therapeutic strategy in a clinical setting.


Subject(s)
Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA, Antisense/pharmacology , Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Cell Division/drug effects , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood supply , Neoplasms/pathology , Oligonucleotides/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 7(5): 1459-65, 2001 May.
Article in English | MEDLINE | ID: mdl-11350918

ABSTRACT

The transforming growth factor-alpha/epidermal growth factor receptor (TGF-alpha-EGFR) autocrine pathway, which is involved in the development and the progression of human epithelial cancers, controls, in part, the production of angiogenic factors. These angiogenic factors, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), are secreted by cancer cells to stimulate normal endothelial cell growth through paracrine mechanisms. ZD1839 (Iressa) is a p.o.-active, selective EGFR-tyrosine kinase inhibitor (TKI) in clinical trials in cancer patients. In this study, we evaluated the antiangiogenic and antitumor activity of ZD1839 in human colon (GEO, SW480, and CaCo2), breast (ZR-75-1 and MCF-7 ADR), ovarian (OVCAR-3), and gastric (KATO III and N87) cancer cells that coexpress TGF-alpha and EGFR. ZD1839 treatment determined a dose- and time-dependent growth inhibition accompanied by the decrease of VEGF, bFGF and TGF-alpha production in vitro. Treatment of immunodeficient mice bearing well-established, palpable GEO xenografts with ZD1839 determined a cytostatic dose-dependent tumor growth inhibition. Immunohistochemical analysis of GEO tumor xenografts after ZD1839 treatment revealed a significant dose-dependent reduction of TGF-alpha, bFGF, and VEGF expression in cancer cells and of neoangiogenesis, as determined by microvessel count. Furthermore, the antitumor activity of ZD1839 was potentiated in combination with the cytotoxic drug paclitaxel in GEO tumor xenografts. Tumor regression was observed in all mice after treatment with ZD1839 plus paclitaxel, and it was accompanied by a significant potentiation in inhibition of TGF-alpha, VEGF, and bFGF expression with a few or no microvessels. Furthermore, 6 of 16 mice bearing well-established, palpable GEO xenografts had no histological evidence of GEO tumors at the end of treatment with ZD1839 plus paclitaxel. These results demonstrate that the antitumor effect of ZD1839 is accompanied by inhibition in the production of autocrine and paracrine growth factors that sustain autonomous local growth and facilitate angiogenesis, and that this effect can be potentiated by the combined treatment with certain cytotoxic drugs, such as paclitaxel.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/metabolism , Growth Substances/metabolism , Neovascularization, Pathologic/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Drug Synergism , Endothelial Growth Factors/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Gefitinib , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Lymphokines/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Protein-Tyrosine Kinases/metabolism , Quinazolines/therapeutic use , Transforming Growth Factor alpha/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 6(11): 4343-50, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106252

ABSTRACT

Recent studies have suggested that selective inhibition of mitogenic pathways may improve the antitumor activity of ionizing radiation. The epidermal growth factor receptor (EGFR) is overexpressed and is involved in autocrine growth control in the majority of human carcinomas. Protein kinase A type I (PKAI) plays a key role in neoplastic transformation and is overexpressed in cancer cells in which an EGFR autocrine pathway is activated. We used two specific inhibitors of EGFR and PKAI that are under clinical evaluation in cancer patients: C225, an anti-EGFR chimeric human-mouse monoclonal antibody (MAb); and a mixed-backbone antisense oligonucleotide targeting the PKAI RIalpha subunit (PKAI AS). We tested in human colon cancer (GEO) and ovarian cancer (OVCAR-3) cell lines the antiproliferative activity of MAb C225 and/or PKAI AS in combination with ionizing radiation. In vivo antitumor activity was evaluated in nude mice bearing established GEO xenografts. Dose-dependent inhibition of soft agar growth was observed in both cancer cell lines with ionizing radiation, C225, or PKAI AS oligonucleotide. A cooperative antiproliferative effect was obtained when cancer cells were treated with ionizing radiation followed by MAb C225 or PKAI AS oligonucleotide. This effect was observed at all doses tested in both GEO and OVCAR-3 cancer cell lines. A combination of the three treatments at the lowest doses produced an even greater effect than that observed when two modalities were combined. Treatment of mice bearing established human GEO colon cancer xenografts with radiotherapy (RT), MAb C225, or PKAI AS oligonucleotide produced dose-dependent tumor growth inhibition that was reversible upon treatment cessation. A potentiation of the antitumor activity was observed in all mice treated with RT in combination with MAb C225 or PKAI AS oligonucleotide. Long-term GEO tumor growth regression was obtained following treatment with ionizing radiation in combination with MAb C225 plus PKAI AS oligonucleotide, which produced a significant improvement in survival compared with controls (P < 0.001), the RT-treated group (P < 0.001), or the group treated with MAb C225 plus PKAI AS oligonucleotide (P < 0.001). All mice of the RT + MAb C225 + PKAI AS group were alive 26 weeks after tumor cell injection. Furthermore, 50% of mice in this group were alive and tumor-free after 35 weeks. This study provides a rationale for evaluating in cancer patients the combination of ionizing radiation and selective drugs that block EGFR and PKAI pathways.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Neoplasms/therapy , Oligonucleotides, Antisense/therapeutic use , Animals , Combined Modality Therapy , Female , Humans , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
19.
Int J Cancer ; 88(5): 804-9, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11072252

ABSTRACT

MDM2 is over-expressed in several human tumors. Its product is a negative-feedback regulator of p53, which interferes with the control of cell proliferation and apoptosis, interacting not only with p53 but also with retinoblastoma (Rb) and E2F. Moreover, mutations in the ARF-Ink4a locus may also allow MDM2 to override p53 functions. In this study, we have used a novel oligonucleotide anti-sense MDM2, with mixed-backbone structure and demonstrate that it causes inhibition of MDM2 expression, induction of both p53 and p21/WAF1 expression and a dose-dependent, growth-inhibitory effect in human GEO colon-cancer cells. We also show that anti-sense MDM2 has a co-operative growth-inhibitory effect with different classes of cytotoxic drugs acting by different mechanisms. Moreover, anti-sense MDM2 induces apoptosis and markedly enhances the apoptotic activity of different cytotoxic drugs. Finally, we show that anti-sense MDM2 has anti-tumor activity in vivo in nude mice bearing GEO xenografts and potentiates the anti-tumor effect of cytotoxic drugs. Indeed, despite the short treatment period, the combination of anti-sense MDM2 and cytotoxic drugs causes a marked delay in tumor growth and prolongation of mice survival, lasting several months after treatment cessation. The anti-tumor effect is associated with inhibition of MDM2 expression in tumor specimens of animals treated with anti-sense MDM2, alone or in combination with a cytotoxic drug. Our results provide the rationale for development of a novel mixed-backbone anti-sense MDM2 into a clinical setting in therapeutic combination strategies with conventional cytotoxic drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Nuclear Proteins , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Division/drug effects , Disease Models, Animal , Drug Synergism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured
20.
Clin Cancer Res ; 6(9): 3739-47, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10999768

ABSTRACT

Angiogenesis plays a key role in tumor growth and metastasis. The transforming growth factor alpha (TGF-alpha)-epidermal growth factor receptor (EGFR) autocrine pathway controls in part the production of angiogenic factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cancer cells. In this study, we have evaluated the antiangiogenic and antitumor activity of monoclonal antibody (MAb) C225, an anti-EGFR chimeric human-mouse MAb, alone and in combination with a human VEGF antisense (AS) 21-mer phosphorothioate oligonucleotide (VEGF-AS) in human GEO colon cancer cells. MAb C225 treatment determined a dose-dependent inhibition of VEGF, bFGF, and TGF-alpha production by GEO cells in vitro. Treatment with VEGF-AS caused a selective inhibition in VEGF expression by GEO cells in vitro. Treatment of immunodeficient mice bearing established, palpable GEO xenografts for 3 weeks with VEGF-AS or with MAb C225 determined a cytostatic reversible inhibition of tumor growth. In contrast, a prolonged inhibition of tumor growth was observed in all mice treated with the two agents, in combination with a significant improvement in mice survival compared with controls (P < .001), to MAb C225 (P < .001), or to VEGF-AS (P < .001) treated mice. All mice died within 4, 6, and 8 weeks after tumor cell injection in the control, VEGF-AS and MAb C225 groups, respectively. In contrast, 50% of mice treated with the combination of VEGF-AS and MAb C225 were alive at 13 weeks. Ten % of mice treated with VEGF-AS plus MAb C225 were alive at 20 weeks and had no histological evidence of GEO tumors. Immunohistochemical analysis of GEO tumor xenografts demonstrated a significant reduction of VEGF expression after treatment with VEGF-AS with a parallel reduction in microvessel count. MAb C225 treatment determined a reduction in the expression of VEGF, bFGF, and TGF-alpha with a reduction in microvessel count. Finally, a significant potentiation in inhibition of VEGF expression and little or no microvessels were observed in GEO tumors after the combined treatment with the two agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/therapy , Endothelial Growth Factors/genetics , Lymphokines/genetics , Neovascularization, Pathologic/therapy , Oligonucleotides, Antisense/pharmacology , Thionucleotides/pharmacology , Animals , Blotting, Western , Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/biosynthesis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Female , Fibroblast Growth Factor 2/biosynthesis , Humans , Immunohistochemistry , Lymphokines/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Transforming Growth Factor alpha/biosynthesis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...