Giovanna Damia
University of Cincinnati
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Giovanna Damia.
European Journal of Cancer | 2001
Eugenio Erba; Daniele Bergamaschi; L Bassano; Giovanna Damia; Simona Ronzoni; G Faircloth; Maurizio D'Incalci
The mode of action of Ecteinascidin-743 (ET-743), a marine tetrahydroisoquinoline alkaloid isolated from Ecteinascidia turbinata, which has shown very potent antitumour activity in preclinical systems and encouraging results in Phase I clinical trials was investigated at a cellular level. Both SW620 and LoVo human intestinal carcinoma cell lines exposed for 1 h to ET-743 progress through S phase more slowly than control cells and then accumulate in the G2M phase. The sensitivity to ET-743 of G1 synchronised cells was much higher than that of cells synchronised in S phase and even higher than that of cells synchronised in G2M. ET-743 concentrations up to four times higher than the IC(50) value caused no detectable DNA breaks or DNA-protein cross-links as assessed by alkaline elution techniques. ET-743 induced a significant increase in p53 levels in cell lines expressing wild-type (wt) (p53). However, the p53 status does not appear to be related to the ET-743 cytotoxic activity as demonstrated by comparing the drug sensitivity in p53 (-/-) or (+/+) mouse embryo fibroblasts and in A2780 ovarian cancer cells or the A2780/CX3 sub-line transfected with a dominant-negative mutant TP53. The cytotoxic potency of ET-743 was comparatively evaluated in CHO cell lines proficient or deficient in nucleotide excision repair (NER), and it was found that ET-743 was approximately 7-8 times less active in ERCC3/XPB and ERCC1-deficient cells than control cells. The findings that G1 phase cells are hypersensitive and that NER-deficient cells are resistant to ET-743 indicate that the mode of action of ET-743 is unique and different from that of other DNA-interacting drugs.
Cancer Treatment Reviews | 2012
Chiara Foroni; Massimo Broggini; Daniele Generali; Giovanna Damia
Epithelial-mesenchymal transition (EMT) is defined by the loss of epithelial characteristics and the acquisition of a mesenchymal phenotype. In this process, cells acquire molecular alterations that facilitate dysfunctional cell-cell adhesive interactions and junctions. These processes may promote cancer cell progression and invasion into the surrounding microenvironment. Such transformation has implications in progression of breast carcinoma to metastasis, and increasing evidences support most tumors contain a subpopulation of cells with stem-like and mesenchymal features that is resistant to chemotherapy. This review focuses on the physiological and pathological role of EMT process, its molecular related network, its putative role in the metastatic process and its implications in response/resistance to the current and/or new approaching drugs in the clinical management of breast cancer.
International Journal of Cancer | 2001
Giovanna Damia; Simonetta Silvestri; Laura Carrassa; Laura Filiberti; Glynn Faircloth; Giordano Liberi; Marco Foiani; Maurizio D'Incalci
The cytotoxic activity of ecteinascidin 743 (ET‐743), a natural product derived from the marine tunicate Ecteinascidia turbinata that exhibits potent anti‐tumor activity in pre‐clinical systems and promising activity in phase I and II clinical trials, was investigated in a number of cell systems with well‐defined deficiencies in DNA‐repair mechanisms. ET‐743 binds to N2 of guanine in the minor groove, but its activity does not appear to be related to DNA‐topoisomerase I poisoning as the drug is equally active in wild‐type yeast and in yeast with a deletion in the DNA‐topoisomerase I gene. Defects in the mismatch repair pathway, usually associated with increased resistance to methylating agents and cisplatin, did not affect the cytotoxic activity of ET‐743. However, ET‐743 did show decreased activity (from 2‐ to 8‐fold) in nucleotide excision repair (NER)–deficient cell lines compared to NER‐proficient cell lines, from either hamsters or humans. Restoration of NER function sensitized cells to ET‐743 treatment. The DNA double‐strand‐break repair pathway was also investigated using human glioblastoma cell lines MO59K and MO59J, respectively, proficient and deficient in DNA‐dependent protein kinase (DNA‐PK). ET‐743 was more effective in cells lacking DNA‐PK; moreover, pre‐treatment of HCT‐116 colon carcinoma cells with wortmannin, a potent inhibitor of DNA‐PK, sensitized cells to ET‐743. An increase in ET‐743 sensitivity was also observed in ataxia telangiectasia–mutated cells. Our data strongly suggest that ET‐743 has a unique mechanism of interaction with DNA.
Cancer Research | 2011
Ezia Bello; Gennaro Colella; Valentina Scarlato; Paolo Oliva; Alexander Berndt; Giovanni Valbusa; Sonia Colombo Serra; Maurizio D'Incalci; Ennio Cavalletti; Raffaella Giavazzi; Giovanna Damia; Gabriella Camboni
Tumor angiogenesis is a degenerate process regulated by a complex network of proangiogenic factors. Existing antiangiogenic drugs used in clinic are characterized by selectivity for specific factors. Antiangiogenic properties might be improved in drugs that target multiple factors and thereby address the inherent mechanistic degeneracy in angiogenesis. Vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) family members and their cognate receptors are key players in promoting tumor angiogenesis. Here we report the pharmacologic profile of E-3810, a novel dual inhibitor of the VEGF and FGF receptors. E-3810 potently and selectively inhibited VEGF receptor (VEGFR)-1, -2, and -3 and FGF receptor (FGFR)-1 and -2 kinases in the nanomolar range. Ligand-dependent phosphorylation of VEGFR-2 and FGFR-1 was suppressed along with human vascular endothelial cell growth at nanomolar concentrations. In contrast, E-3810 lacked cytotoxic effects on cancer cell lines under millimolar concentrations. In a variety of tumor xenograft models, including early- or late-stage subcutaneous and orthotopic models, E-3810 exhibited striking antitumor properties at well-tolerated oral doses administered daily. We found that E-3810 remained active in tumors rendered nonresponsive to the general kinase inhibitor sunitinib resulting from a previous cycle of sunitinib treatment. In Matrigel plug assays performed in nude mice, E-3810 inhibited basic FGF-induced angiogenesis and reduced blood vessel density as assessed by histologic analysis. Dynamic contrast-enhanced magnetic resonance imaging analysis confirmed that E-3810 reduced the distribution of angiogenesis-sensitive contrast agents after only 5 days of treatment. Taken together, our findings identify E-3810 as a potent antiangiogenic small molecule with a favorable pharmacokinetic profile and broad spectrum antitumor activity, providing a strong rationale for its clinical evaluation.
International Journal of Cancer | 1996
Giovanna Damia; Luigi Imperatori; Miria Stefanini; Maurizio D'Incalci
Nucleotide excision repair (NER) is one of the major DNA repair systems in mammalian cells, able to remove a broad spectrum of unrelated lesions. In this report the role of ERCC (excision repair cross‐complementing) 1, ERCC2, ERCC3, ERCC4 and ERCC6 genes in removing the lesions caused by alkylating agents with different structures and mechanisms of action has been studied using UV‐sensitive DNA repair‐deficient mutant CHO cell lines. We confirmed that ERCC1 and ERCC4 play a role in the repair of cis‐diamminedichloroplatinum (DDP)‐ and Melphalan (L‐PAM)‐induced DNA damage, while a marginal role of ERCC2 and ERCC3 in the cellular response to DDP and L‐PAM treatment has been observed. Treatment with methylating agents (DM and MNNG) showed a lack of a preferential cytotoxicity between the parental and the different NER‐deficient cell lines, emphasizing the importance of other repair systems such as 3‐methyladenine glycosylase and O6 alkyl‐guanine‐DNA‐alkyl‐transferase. ERCC1, ERCC2, ERCC3 and ERCC4, but not ERCC6 gene products seem to be involved in removing the lesions caused by Tallimustine and CC1065, minor groove alkylating agents that alkylate N3 adenine in a sequence‐specific manner. ERCC6‐deficient cells were as sensitive as the parental cell line to all the cytotoxic drugs tested, except DDP. These data emphasize the importance of the CHO mutant cell lines with specific defects in the DNA repair system for investigating the mechanism of action of different anti‐cancer agents.
European Journal of Cancer | 2009
Giovanna Damia; Maurizio D’Incalci
The successful identification of novel effective anticancer drugs is largely dependent on the use of appropriate preclinical experimental models that should possibly mimic the complexity of different cancer diseases. The huge number of targets suitable for the design of new anticancer drugs is producing hundreds of novel molecules that require appropriate experimental models to investigate their mode of action and antitumour activity in order to select for clinical investigation the ones with higher chances of being clinically effective. However, our ability to predict the clinical efficacy of a new compound in the clinic based on preclinical data is still limited. This paper overviews the in vitro/in vivo preclinical systems that are currently used to test either compounds with an unknown mechanism of action or compounds designed to hit cancer-specific or cancer-related molecular targets. Examples of experimental models successfully used to identify novel compounds are provided. Xenografts are still the most commonly used in vivo models in drug development due to their high degree of reproducibility and because, in some cases, particularly when orthotopically transplanted, they maintain several biological properties of the human tumours they derive from. Genetic models are very useful for target validation, but are often not sufficiently reproducible to be used for drug evaluation. The variety of animal models can be effectively used to optimally test drugs that presumably act by a defined mode of action, but final success is highly dependent on the ability of drug development teams to integrate different expertises such as biology, chemistry, pharmacology, toxicology and clinical oncology into a clever and well orchestrated plan that keeps in consideration both the complexity of cancer diseases, involving alterations of different pathways, and the complexity of drugs whose pharmacological properties are crucial to obtain the desired effects.
European Journal of Cancer | 2008
Michele Tavecchio; Matteo Simone; Eugenio Erba; Irene Chiolo; Giordano Liberi; Marco Foiani; M. D’Incalci; Giovanna Damia
Trabectedin (ET-743, Yondelis) is a natural marine compound with antitumour activity currently undergoing phase II/III clinical trials. The mechanism of the drugs action is still to be defined, even though it has been clearly demonstrated the key role of Nucleotide Excision Repair (NER). To get further insights into the drugs mode of action, we studied the involvement of the DNA-double strand break repair (DNA-DSB) pathways: homologous and non-homologous recombination, both in budding yeasts and in mammalian cells and the possible cross-talk between NER and these repair pathways. Budding yeasts and mammalian cells deficient in the non-homologous end-joining pathway were moderately sensitive to trabectedin, while systems deficient in the homologous recombination pathway were extremely sensitive to the drug, with a 100-fold decrease in the IC50, suggesting that trabectedin-induced lesions are repaired by this pathway. The induction of Rad51 foci and the appearance of gamma-H2AX were chosen as putative markers for DNA-DSBs and were studied at different time points after trabectedin treatment in NER proficient and deficient systems. Both were clearly detected only in the presence of an active NER, suggesting that the DSBs are not directly caused by the drug, but are formed during the processing/repair of the drug- induced lesions.
Cell Cycle | 2012
Laura Carrassa; Rosaria Chilà; Monica Lupi; Francesca Ricci; Cinzia Celenza; Marco Mazzoletti; Massimo Broggini; Giovanna Damia
Targeting Chk1 protein kinase can enhance the antitumor effects of radio- and chemotherapy. Recent evidence disclosed a role of Chk1 in unperturbed cell proliferation and survival, implying that Chk1 inhibitors could also be effective as single agents in tumors with a specific genetic background. To identify genes in synthetic lethality with Chk1, we did a high-throughput screening using a siRNA library directed against 719 human protein kinases in the human ovarian cancer cell line OVCAR-5, resistant to Chk1 inhibitors. Wee1 tyrosine kinase was the most significant gene in synthetic lethality with Chk1. Treatment with non-toxic concentrations of a Chk1 inhibitor (PF-00477736) and a Wee1 inhibitor (MK-1775) confirmed the marked synergistic effect in various human cancer cell lines (breast, ovarian, colon, prostate), independently of the p53 status. Detailed molecular analysis showed that the combination caused cancer cells to undergo premature mitosis before the end of DNA replication, with damaged DNA leading to cell death partly by apoptosis. In vivo treatment of mice bearing OVCAR-5 xenografts with the combination of Chk1 and Wee1 inhibitors led to greater tumor growth inhibition than with the inhibitors used as single agents with no toxicity. These data provide a strong rationale for the clinical investigation of the combination of a Chk1 and a Wee1 inhibitor.
Cell Cycle | 2004
Laura Carrassa; Massimo Broggini; Eugenio Erba; Giovanna Damia
Mammalian Chk1 and Chk2 protein kinases are two important components of the G2DNA damage checkpoint. They are activated by upstream kinases (ataxia telangectasiamutated gene and ATM and Rad 3 related gene) and interfere with the activity of thecdc2/cyclinB1 complex, necessary for the G2-M transition, through the inactivation of thecdc25 phosphatases (cdc25A and cdc25C). To understand the role of Chk1 and Chk2 in thecellular response to different anticancer agents, we knocked down the expression of eachprotein or simultaneously of both proteins by using the small interfering RNA techniquein the HCT-116 colon carcinoma cell line and in its isogenic systems in which p53 and p21have been inactivated by targeted homologous recombination. We here show thatinhibition of Chk1 but not of Chk2 in p21-/- and p53-/- cells caused a greater abrogationof G2 block induced by ionizing radiation and cis-diamine-dichloroplatinum treatmentsand a greater sensitisation to the same treatments than in the parental cell line with p53and p21 wild type proteins. These data further emphasise the role of Chk1 as a moleculartarget to inhibit in tumors with a defect in the G1 checkpoint with the aim of increasingthe selectivity and specificity of anticancer drug treatments.
Cell Cycle | 2011
Laura Carrassa; Giovanna Damia
The checkpoint kinase 1 (Chk1) is one of the major players in the signal transduction pathway set in motion in response to DNA damage which activates different cell cycle checkpoints including the G1/S, the intra-S, G2-M and the mitotic spindle checkpoint, contributing to the maintenance of genomic stability. Chk1 is considered a good molecular target to inhibit, in combination with other anticancer agents, to increase the sensitivity of treatment, especially in tumors with a defective G1 checkpoint. Experimental evidence highlights the essential role of Chk1 in normal and cancer cells even under unstressed conditions, especially in controlling DNA replication and cell division. This review looks at the main functions of Chk1 and the data on Chk1 inhibitors at their preclinical and clinical development are reported. This information may suggest novel approaches for new treatments with Chk1 inhibitors in combination with anticancer agents or as single agents. The emergent synthetic lethality approach may help define the genetic background features where Chk1 inhibitors alone could be very effective.